Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Epidemiol Infect ; 151: e126, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37469288

RESUMEN

This work aimed to study the role of different SARS-CoV-2 lineages in the epidemiology of multiple waves of the COVID-19 pandemic in Ribeirão Preto (São Paulo state), with comparison within Brazil and globally. Viral genomic sequencing was combined with clinical and sociodemographic information of 2,379 subjects at a large Brazilian hospital. On the whole 2,395 complete SARS-CoV-2 genomes were obtained from April 2020 to January 2022. We report variants of concern (VOC) and interest (VOI) dynamics and the role of Brazilian lineages. We identified three World Health Organization VOCs (Gamma, Delta, Omicron) and one VOI (Zeta), which caused distinct waves in this cohort. We also identified 47 distinct Pango lineages. Consistent with the high prevalence of Gamma in Brazil, Pango lineage P.1 dominated infections in this cohort for half of 2021. Each wave of infection largely consisted of a single variant group, with each new group quickly and completely rising to dominance. Despite increasing vaccination in Brazil starting in 2021, this pattern was observed throughout the study and is consistent with the hypothesis that herd immunity tends to be SARS-CoV-2 variant-specific and does not broadly protect against COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Brasil/epidemiología , Pandemias , COVID-19/epidemiología , Genómica , Hospitales Universitarios
2.
J Clin Endocrinol Metab ; 108(6): 1452-1463, 2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-36504388

RESUMEN

OBJECTIVE: To explore pituitary tumors by methylome and transcriptome signatures in a heterogeneous ethnic population. METHODS: In this retrospective cross-sectional study, clinicopathological features, methylome, and transcriptome were evaluated in pituitary tumors from 77 patients (61% women, age 12-72 years) followed due to functioning (FPT: GH-secreting n = 18, ACTH-secreting n = 14) and nonfunctioning pituitary tumors (NFPT, n = 45) at Ribeirao Preto Medical School, University of São Paulo. RESULTS: Unsupervised hierarchical clustering analysis (UHCA) of methylome (n = 77) and transcriptome (n = 65 out of 77) revealed 3 clusters each: one enriched by FPT, one by NFPT, and a third by ACTH-secreting and NFPT. Comparison between each omics-derived clusters identified 3568 and 5994 differentially methylated and expressed genes, respectively, which were associated with each other, with tumor clinical presentation, and with 2017 and 2022 WHO classifications. UHCA considering 11 transcripts related to pituitary development/differentiation also supported 3 clusters: POU1F1-driven somatotroph, TBX19-driven corticotroph, and NR5A1-driven gonadotroph adenomas, with rare exceptions (NR5A1 expressed in few GH-secreting and corticotroph silent adenomas; POU1F1 in few ACTH-secreting adenomas; and TBX19 in few NFPTs). CONCLUSION: This large heterogenic ethnic Brazilian cohort confirms that integrated methylome and transcriptome signatures classify FPT and NFPT, which are associated with clinical presentation and tumor invasiveness. Moreover, the cluster NFPT/ACTH-secreting adenomas raises interest regarding tumor heterogeneity, supporting the challenge raised by the 2017 and 2022 WHO definition regarding the discrepancy, in rare cases, between clinical presentation and pituitary lineage markers. Finally, making our data publicly available enables further studies to validate genes/pathways involved in pituitary tumor pathogenesis and prognosis.


Asunto(s)
Adenoma Hipofisario Secretor de ACTH , Adenoma , Neoplasias Hipofisarias , Humanos , Femenino , Niño , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Masculino , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Adenoma/genética , Adenoma/patología , Epigenoma , Transcriptoma , Estudios Retrospectivos , Estudios Transversales , Adenoma Hipofisario Secretor de ACTH/genética , Hormona Adrenocorticotrópica/genética
3.
Arch. Head Neck Surg ; 51: e20220004, Jan-Dec. 2022.
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1401105

RESUMEN

Introduction: Head and neck cancer (HNC) is a heterogeneous group of neoplasms that can have a poor prognosis when diagnosed in advanced stages. The optimized treatment for locally advanced and unresectable lesions is mainly based on radiotherapy associated with chemotherapy (cisplatin 100mg/m²), however, at the expense of a high toxicity index. Objective: Evaluate whether chemoradiotherapy (CRT) ­ the goldstandard treatment for locally advanced head and neck cancer (HNC) ­ is effective in the study population. Methods: This is a retrospective study aimed at determining the efficacy of definitive CRT in patients with unresectable HNC treated between the 2012 and 2018 in a single institution. The following outcomes were evaluated: objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and toxicity profiles. Results: Fifty-two (52) patients diagnosed with HNC between 2012 and 2018 met the inclusion criteria. The ORR was 84.6%, with 50% showing complete response. Median PFS and OS were 35.3 and 52 months, respectively. Analysis of the toxicity profiles revealed that 69.2% of the patients presented grade 3-4 toxicity. Completion of two or more cycles of cisplatin-based therapy (HR 3.57 [95% CI 1.25­10.25]; p p<0.001), grade 3-4 toxicity (HR 0.27 [95% CI 0.09-0.8] ­ p<0.02), and Charlson comorbidity index (CCI) (HR 3.23 [95% CI 1.26­8.29]; p<0.001) were significantly associated with survival. Regarding toxicity, prophylactic low-level laser therapy (HR 0.48 [95% CI 0.27­0.86]; p<0.001 for those without this practice) and body mass index (BMI) (HR 0.27 [95% CI 0.09­0.76]; p<0.01) showed statistical significance. Conclusion: CRT was effective to treat HNC in the study population, with PFS and OS comparable to those reported in larger sample studies and lower toxicity grade. Some clinical characteristics have been identified as prognostic and/or predictive factors.

4.
J Immunother Cancer ; 10(10)2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36220303

RESUMEN

BACKGROUND: Colorectal cancer (CRC) has a high mortality rate and can develop in either colitis-dependent (colitis-associated (CA)-CRC) or colitis-independent (sporadic (s)CRC) manner. There has been a significant debate about whether mast cells (MCs) promote or inhibit the development of CRC. Herein we investigated MC activity throughout the multistepped development of CRC in both human patients and animal models. METHODS: We analyzed human patient matched samples of healthy colon vs CRC tissue alongside conducting a The Cancer Genome Atlas-based immunogenomic analysis and multiple experiments employing genetically engineered mouse (GEM) models. RESULTS: Analyzing human CRC samples revealed that MCs can be active or inactive in this disease. An activated MC population decreased the number of tumor-residing CD8 T cells. In mice, MC deficiency decreased the development of CA-CRC lesions, while it increased the density of tumor-based CD8 infiltration. Furthermore, co-culture experiments revealed that tumor-primed MCs promote apoptosis in CRC cells. In MC-deficient mice, we found that MCs inhibited the development of sCRC lesions. Further exploration of this with several GEM models confirmed that different immune responses alter and are altered by MC activity, which directly alters colon tumorigenesis. Since rescuing MC activity with bone marrow transplantation in MC-deficient mice or pharmacologically inhibiting MC effects impacts the development of sCRC lesions, we explored its therapeutic potential against CRC. MC activity promoted CRC cell engraftment by inhibiting CD8+ cell infiltration in tumors, pharmacologically blocking it inhibits the ability of allograft tumors to develop. This therapeutic strategy potentiated the cytotoxic activity of fluorouracil chemotherapy. CONCLUSION: Therefore, we suggest that MCs have a dual role throughout CRC development and are potential druggable targets against this disease.


Asunto(s)
Colitis , Neoplasias Colorrectales , Animales , Fluorouracilo , Humanos , Mastocitos , Ratones
5.
Nat Genet ; 54(8): 1103-1116, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35835913

RESUMEN

The chr12q24.13 locus encoding OAS1-OAS3 antiviral proteins has been associated with coronavirus disease 2019 (COVID-19) susceptibility. Here, we report genetic, functional and clinical insights into this locus in relation to COVID-19 severity. In our analysis of patients of European (n = 2,249) and African (n = 835) ancestries with hospitalized versus nonhospitalized COVID-19, the risk of hospitalized disease was associated with a common OAS1 haplotype, which was also associated with reduced severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) clearance in a clinical trial with pegIFN-λ1. Bioinformatic analyses and in vitro studies reveal the functional contribution of two associated OAS1 exonic variants comprising the risk haplotype. Derived human-specific alleles rs10774671-A and rs1131454 -A decrease OAS1 protein abundance through allele-specific regulation of splicing and nonsense-mediated decay (NMD). We conclude that decreased OAS1 expression due to a common haplotype contributes to COVID-19 severity. Our results provide insight into molecular mechanisms through which early treatment with interferons could accelerate SARS-CoV-2 clearance and mitigate against severe COVID-19.


Asunto(s)
COVID-19 , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Alelos , COVID-19/genética , Hospitalización , Humanos , SARS-CoV-2/genética
6.
Eur J Endocrinol ; 187(2): 219-230, 2022 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-35584004

RESUMEN

Objectives: To evaluate how telomere length behaves in adamantinomtous craniopharyngioma (aCP) and if it contributes to the pathogenesis of aCPs with and without CTNNB1 mutations. Design: Retrospective cross-sectional study enrolling 42 aCP patients from 2 tertiary institutions. Methods: Clinicopathological features were retrieved from the patient's charts. Fresh frozen tumors were used for RNA and DNA analyses. Telomere length was evaluated by qPCR (T/S ratio). Somatic mutations in TERT promoter (TERTp) and CTNNB1 were detected by Sanger and/or whole-exome sequencing. We performed RNA-Seq to identify differentially expressed genes in aCPs presenting with shorter or longer telomere lengths. Results: Mutations in CTNNB1 were detected in 29 (69%) tumors. There was higher frequency of CTNNB1 mutations in aCPs from patients diagnosed under the age of 15 years (85% vs 15%; P = 0.04) and a trend to recurrent disease (76% vs 24%; P = 0.1). No mutation was detected in the TERTp region. The telomeres were shorter in CTNNB1-mutated aCPs (0.441, IQR: 0.297-0.597vs 0.607, IQR: 0.445-0.778; P = 0.04), but it was neither associated with clinicopathological features nor with recurrence. RNAseq identified a total of 387 differentially expressed genes, generating two clusters, being one enriched for short telomeres and CTNNB1-mutated aCPs. Conclusions: CTNNB1: mutations are more frequent in children and adolescents and appear to associate with progressive disease. CTNNB1-mutated aCPs have shorter telomeres, demonstrating a relationship between the Wnt/ß-catenin pathway and telomere biology in the pathogenesis of aCPs.


Asunto(s)
Craneofaringioma , Telómero , beta Catenina , Adolescente , Niño , Craneofaringioma/genética , Estudios Transversales , Humanos , Mutación , Estudios Retrospectivos , Telómero/ultraestructura , Vía de Señalización Wnt , beta Catenina/genética
7.
Front Immunol ; 13: 836922, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35386696

RESUMEN

Tumor cells present many strategies for survival and dissemination in the tumor environment. Extracellular vesicles are a vital pathway used in crosstalk between tumor and non-malignant cells. They carry different types of molecules that, when internalized by target cells, can activate signaling pathways and molecular processes that will promote and disseminate neoplastic cells. Proteins, nucleic acids, and different cytokines, such as interleukins, are the main classes of molecules carried by extracellular vesicles and are being studied to understand the molecular mechanisms present in the tumor microenvironment. In particular, although poorly understood, the association between EVs and interleukins has revealed potential approaches to the diagnosis and therapeutics of several neoplasms.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Vesículas Extracelulares/metabolismo , Humanos , Interleucinas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Transducción de Señal , Microambiente Tumoral
8.
Front Immunol ; 13: 826577, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35211121

RESUMEN

Cancer cells harbor genomic instability due to accumulated DNA damage, one of the cancer hallmarks. At least five major DNA Damage Repair (DDR) pathways are recognized to repair DNA damages during different stages of the cell cycle, comprehending base excision repair (BER), nucleotide excision repair (NER), mismatch repair (MMR), homologous recombination (HR), and non-homologous end joining (NHEJ). The unprecedented benefits achieved with immunological checkpoint inhibitors (ICIs) in tumors with mismatch repair deficiency (dMMR) have prompted efforts to extend this efficacy to tumors with HR deficiency (HRD), which are greatly sensitive to chemotherapy or PARP inhibitors, and also considered highly immunogenic. However, an in-depth understanding of HRD's molecular underpinnings has pointed to essential singularities that might impact ICIs sensitivity. Here we address the main molecular aspects of HRD that underlie a differential profile of efficacy and resistance to the treatment with ICIs compared to other DDR deficiencies.


Asunto(s)
Recombinación Homóloga/genética , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Ciclo Celular , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/genética , Reparación de la Incompatibilidad de ADN/genética , Reparación del ADN/genética , Humanos , Neoplasias/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico
9.
Hematol Transfus Cell Ther ; 43 Suppl 2: S78-S83, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34794801

RESUMEN

The use of immunotherapy in cancer treatment over the past decade has resulted in significant advances and improvements in cancer patients survival with the use of checkpoint inhibitors. Nevertheless, only a fraction of solid tumors responds to this immunotherapy modality. Another modality of immunotherapy consists of employing cell-based therapy as an adoptive therapeutic modality. That involves distinct modalities of cellular therapies such as CAR T cells (chimeric antigen receptor T cell), TILs (tumor-infiltrating lymphocytes), and TCR T cells. Those treatments have proven effective in hematologic tumors and could have an impact in tumors that do not respond to checkpoint inhibitors. This review aims to outline the rationale, operation, clinical applicability, and results of adoptive cell therapy for patients with solid tumors.

10.
Am J Hum Genet ; 108(9): 1590-1610, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34390653

RESUMEN

Our study investigated the underlying mechanism for the 14q24 renal cell carcinoma (RCC) susceptibility risk locus identified by a genome-wide association study (GWAS). The sentinel single-nucleotide polymorphism (SNP), rs4903064, at 14q24 confers an allele-specific effect on expression of the double PHD fingers 3 (DPF3) of the BAF SWI/SNF complex as assessed by massively parallel reporter assay, confirmatory luciferase assays, and eQTL analyses. Overexpression of DPF3 in renal cell lines increases growth rates and alters chromatin accessibility and gene expression, leading to inhibition of apoptosis and activation of oncogenic pathways. siRNA interference of multiple DPF3-deregulated genes reduces growth. Our results indicate that germline variation in DPF3, a component of the BAF complex, part of the SWI/SNF complexes, can lead to reduced apoptosis and activation of the STAT3 pathway, both critical in RCC carcinogenesis. In addition, we show that altered DPF3 expression in the 14q24 RCC locus could influence the effectiveness of immunotherapy treatment for RCC by regulating tumor cytokine secretion and immune cell activation.


Asunto(s)
Carcinoma de Células Renales/genética , Cromosomas Humanos Par 14 , Proteínas de Unión al ADN/genética , Sitios Genéticos , Neoplasias Renales/genética , Factor de Transcripción STAT3/genética , Factores de Transcripción/genética , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/patología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/terapia , Línea Celular Tumoral , Cromatina/química , Cromatina/inmunología , Ensamble y Desensamble de Cromatina/inmunología , Citocinas/genética , Citocinas/inmunología , Proteínas de Unión al ADN/inmunología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genoma Humano , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunoterapia/métodos , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Neoplasias Renales/terapia , Polimorfismo de Nucleótido Simple , Factor de Transcripción STAT3/inmunología , Linfocitos T Citotóxicos , Factores de Transcripción/inmunología
11.
Science ; 372(6543): 725-729, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33888597

RESUMEN

Effects of radiation exposure from the Chernobyl nuclear accident remain a topic of interest. We investigated germline de novo mutations (DNMs) in children born to parents employed as cleanup workers or exposed to occupational and environmental ionizing radiation after the accident. Whole-genome sequencing of 130 children (born 1987-2002) and their parents did not reveal an increase in the rates, distributions, or types of DNMs relative to the results of previous studies. We find no elevation in total DNMs, regardless of cumulative preconception gonadal paternal [mean = 365 milligrays (mGy), range = 0 to 4080 mGy] or maternal (mean = 19 mGy, range = 0 to 550 mGy) exposure to ionizing radiation. Thus, we conclude that, over this exposure range, evidence is lacking for a substantial effect on germline DNMs in humans, suggesting minimal impact from transgenerational genetic effects.

12.
Transl Oncol ; 14(1): 100970, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33260070

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC) remains a challenging cancer to treat despite all the advances of the last 50 years. Kallikrein 5 (KLK5) is among the serine proteases implicated in OSCC development. However, whether the activity of KLK5 promotes carcinogenesis is still controversial. Moreover, knowledge regarding the role of the KLK5 cognate inhibitor, Lympho-Epithelial Kazal-Type related Inhibitor (LEKTI), in OSCC is scarce. We have, thus, sought to investigate the importance of KLK5 and LEKTI expression in premalignant and malignant lesions of the oral cavity. METHODS: KLK5 and LEKTI protein expression was evaluated in 301 human samples, which were comprised of non-malignant and malignant lesions of the oral cavity. Moreover, a bioinformatic analysis of the overall survival rate from 517 head and neck squamous cell carcinoma (HNSCC) samples was performed. Additionally, to mimic the uncovered KLK5 to serine peptidase inhibitor (SPINK5) imbalance, the KLK5 gene was abrogated in an OSCC cell line using CRISPR-Cas9 technology. The generated cell line was then used for in vivo and in vitro carcinogenesis related experiments. RESULTS: LEKTI was found to be statistically downregulated in OSCCs, with increased KLK5/SPINK5 mRNA ratio being associated with a shorter overall survival (p = 0.091). Indeed, disruption of KLK5 to SPINK5 balance through the generation of KLK5 null OSCC cells led to smaller xenografted tumors and statistically decreased proliferation rates following multiple time points of BrdU treatment in vitro. CONCLUSION: The association of increased enzyme/inhibitor ratio with poor prognosis indicates KLK5 to SPINK5 relative expression as an important prognostic marker in OSCC.

13.
Curr Opin Genet Dev ; 66: 1-9, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33307238

RESUMEN

Immune checkpoint inhibitors (ICI) aim to restore the immune system anti-tumor function by blocking two inhibitory axes: CTLA-4/CD28 and PD1/PDL1. ICI is established as a treatment option for multiple cancers, but their remarkable clinical impact is observed only in a fraction of patients. Together with their adverse effects and high cost, it's imperative to identify patients who are likely to benefit from this type of treatment. Genomic features represent promising candidates as predictive biomarkers of response to ICI, with agnostic FDA-approvals of an anti-PD1 drug for tumors with microsatellite instability and tumors with a high mutational burden. Other genomic markers are also emerging to help refine patient selection. In this review, we discuss recent progress in genomic biomarkers development and its challenges, with a focus on alterations in the neoantigen burden, immune, and oncogenic pathways.


Asunto(s)
Biomarcadores de Tumor/genética , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/efectos adversos , Neoplasias/terapia , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Biomarcadores de Tumor/inmunología , Antígenos CD28/antagonistas & inhibidores , Antígenos CD28/genética , Antígenos CD28/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Genómica/tendencias , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología
14.
Sci Rep ; 10(1): 21674, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33303880

RESUMEN

Cell-free DNA is present in different biological fluids and when released by tumor cells may contribute to pro-tumor events such as malignant transformation of cells adjacent to the tumor and metastasis. Thus, this study analyzed the effect of tumor cell-free DNA, isolated from the blood of prostate cancer patients, on non-tumor prostate cell lines (RWPE-1 and PNT-2). To achieve this, we performed cell-free DNA quantification and characterization assays, evaluation of gene and miRNA expression profiling focused on cancer progression and EMT, and metabolomics by mass spectrometry and cellular migration. The results showed that tumor-free cell DNA was able to alter the gene expression of MMP9 and CD44, alter the expression profile of nine miRNAs, and increased the tryptophan consumption and cell migration rates in non-tumor cells. Therefore, tumor cell-free DNA was capable of altering the receptor cell phenotype, triggering events related to malignant transformation in these cells, and can thus be considered a potential target for cancer diagnosis and therapy.


Asunto(s)
Transformación Celular Neoplásica/genética , ADN Tumoral Circulante/efectos adversos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Movimiento Celular/genética , ADN Tumoral Circulante/análisis , ADN Tumoral Circulante/aislamiento & purificación , Progresión de la Enfermedad , Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Próstata/metabolismo , Triptófano/metabolismo
15.
Nat Genet ; 52(12): 1333-1345, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33230299

RESUMEN

Genome-wide association studies identify genomic variants associated with human traits and diseases. Most trait-associated variants are located within cell-type-specific enhancers, but the molecular mechanisms governing phenotypic variation are less well understood. Here, we show that many enhancer variants associated with red blood cell (RBC) traits map to enhancers that are co-bound by lineage-specific master transcription factors (MTFs) and signaling transcription factors (STFs) responsive to extracellular signals. The majority of enhancer variants reside on STF and not MTF motifs, perturbing DNA binding by various STFs (BMP/TGF-ß-directed SMADs or WNT-induced TCFs) and affecting target gene expression. Analyses of engineered human blood cells and expression quantitative trait loci verify that disrupted STF binding leads to altered gene expression. Our results propose that the majority of the RBC-trait-associated variants that reside on transcription-factor-binding sequences fall in STF target sequences, suggesting that the phenotypic variation of RBC traits could stem from altered responsiveness to extracellular stimuli.


Asunto(s)
Eritrocitos/fisiología , Regulación de la Expresión Génica/genética , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Factores de Transcripción/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Eritrocitos/citología , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Sitios de Carácter Cuantitativo/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/genética
16.
Nat Commun ; 11(1): 2718, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32483191

RESUMEN

Genome-wide association studies (GWAS) have identified ~20 melanoma susceptibility loci, most of which are not functionally characterized. Here we report an approach integrating massively-parallel reporter assays (MPRA) with cell-type-specific epigenome and expression quantitative trait loci (eQTL) to identify susceptibility genes/variants from multiple GWAS loci. From 832 high-LD variants, we identify 39 candidate functional variants from 14 loci displaying allelic transcriptional activity, a subset of which corroborates four colocalizing melanocyte cis-eQTL genes. Among these, we further characterize the locus encompassing the HIV-1 restriction gene, MX2 (Chr21q22.3), and validate a functional intronic variant, rs398206. rs398206 mediates the binding of the transcription factor, YY1, to increase MX2 levels, consistent with the cis-eQTL of MX2 in primary human melanocytes. Melanocyte-specific expression of human MX2 in a zebrafish model demonstrates accelerated melanoma formation in a BRAFV600E background. Our integrative approach streamlines GWAS follow-up studies and highlights a pleiotropic function of MX2 in melanoma susceptibility.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Melanoma/genética , Mutación , Proteínas de Resistencia a Mixovirus/genética , Polimorfismo de Nucleótido Simple , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genes Reporteros/genética , Células HEK293 , Humanos , Melanocitos/metabolismo , Melanoma/patología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Sitios de Carácter Cuantitativo/genética , Pez Cebra/genética , Pez Cebra/metabolismo
17.
Sci Rep ; 9(1): 18768, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31822727

RESUMEN

Emerging evidence suggests that dysregulation of oncogenic pathways requires precise tuning in order for cancer to develop. To test this, we examined the overlap between cis-acting elements of the hypoxia-inducible factor (HIF) pathway and cancer-susceptibility polymorphisms as defined in genome-wide association studies (GWAS). In renal cancer, where HIF is constitutively and un-physiologically activated by mutation of the von Hippel-Lindau tumour suppressor, we observed marked excess overlap, which extended to potential susceptibility polymorphisms that are below the conventional threshold applied in GWAS. In contrast, in other cancers where HIF is upregulated by different mechanisms, including micro-environmental hypoxia, we observed no excess in overlap. Our findings support a 'pathway tuning' model of cancer, whereby precise modulation of multiple outputs of specific, activated pathways is important in oncogenesis. This implies that selective pressures to modulate such pathways operate during cancer development and should focus attempts to identify their nature and consequences.


Asunto(s)
Carcinogénesis/genética , Carcinoma de Células Renales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , Transducción de Señal/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/patología , Mutación , Polimorfismo de Nucleótido Simple , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
18.
Eur J Hum Genet ; 27(10): 1589-1598, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31231134

RESUMEN

Renal cell carcinoma (RCC) has an undisputed genetic component and a stable 2:1 male to female sex ratio in its incidence across populations, suggesting possible sexual dimorphism in its genetic susceptibility. We conducted the first sex-specific genome-wide association analysis of RCC for men (3227 cases, 4916 controls) and women (1992 cases, 3095 controls) of European ancestry from two RCC genome-wide scans and replicated the top findings using an additional series of men (2261 cases, 5852 controls) and women (1399 cases, 1575 controls) from two independent cohorts of European origin. Our study confirmed sex-specific associations for two known RCC risk loci at 14q24.2 (DPF3) and 2p21(EPAS1). We also identified two additional suggestive male-specific loci at 6q24.3 (SAMD5, male odds ratio (ORmale) = 0.83 [95% CI = 0.78-0.89], Pmale = 1.71 × 10-8 compared with female odds ratio (ORfemale) = 0.98 [95% CI = 0.90-1.07], Pfemale = 0.68) and 12q23.3 (intergenic, ORmale = 0.75 [95% CI = 0.68-0.83], Pmale = 1.59 × 10-8 compared with ORfemale = 0.93 [95% CI = 0.82-1.06], Pfemale = 0.21) that attained genome-wide significance in the joint meta-analysis. Herein, we provide evidence of sex-specific associations in RCC genetic susceptibility and advocate the necessity of larger genetic and genomic studies to unravel the endogenous causes of sex bias in sexually dimorphic traits and diseases like RCC.


Asunto(s)
Carcinoma de Células Renales/epidemiología , Carcinoma de Células Renales/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Neoplasias Renales/epidemiología , Neoplasias Renales/genética , Biología Computacional , Femenino , Humanos , Masculino , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Factores Sexuales
20.
J Hum Genet ; 64(6): 545-550, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30850729

RESUMEN

Mosaic protein truncating variants (PTVs) in the phosphatase, Mg2+/Mn2+dependent 1D (PPM1D) gene in blood-derived DNA have been associated with increased risk of breast cancer. We analyzed PPM1D PTVs in blood from 3817 breast cancer cases and 3058 controls by deep sequencing of a previously defined region in exon 6 of PPM1D. We identified 50 of 6875 (0.73%) participants having a mosaic PPM1D PTV. We observed a higher frequency of mosaic PPM1D PTVs with increasing age (Ptrend = 2.9 × 10-6). We did not observe an overall association between PPM1D PTVs and increased breast cancer risk (OR = 1.51, 95% CI = 0.84-2.71). Evidence for an association was observed in a subset of cases with DNA collected 1-year or more before breast cancer diagnosis (OR = 3.44, 95% CI = 1.62-7.30, P-value = 0.001); however, no significant association was observed for the larger series of cases with DNA collected post diagnosis (OR = 1.01, 95% CI = 0.51-2.01, P-value = 0.98). Our study indicates that the PPM1D PTVs are present at higher rates than previously reported and the frequency of PPM1D PTVs increases with age. We observed limited evidence for an association between mosaic PPM1D PTVs and breast cancer risk, suggesting mosaic PPM1D PTVs in the blood likely do not influence risk of breast cancer.


Asunto(s)
Envejecimiento/genética , Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad , Proteína Fosfatasa 2C/genética , Anciano , Envejecimiento/patología , Neoplasias de la Mama/patología , Exones , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Persona de Mediana Edad , Mutación , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA