Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nature ; 615(7951): 285-291, 2023 03.
Article in English | MEDLINE | ID: mdl-36859541

ABSTRACT

The germline mutation rate determines the pace of genome evolution and is an evolving parameter itself1. However, little is known about what determines its evolution, as most studies of mutation rates have focused on single species with different methodologies2. Here we quantify germline mutation rates across vertebrates by sequencing and comparing the high-coverage genomes of 151 parent-offspring trios from 68 species of mammals, fishes, birds and reptiles. We show that the per-generation mutation rate varies among species by a factor of 40, with mutation rates being higher for males than for females in mammals and birds, but not in reptiles and fishes. The generation time, age at maturity and species-level fecundity are the key life-history traits affecting this variation among species. Furthermore, species with higher long-term effective population sizes tend to have lower mutation rates per generation, providing support for the drift barrier hypothesis3. The exceptionally high yearly mutation rates of domesticated animals, which have been continually selected on fecundity traits including shorter generation times, further support the importance of generation time in the evolution of mutation rates. Overall, our comparative analysis of pedigree-based mutation rates provides ecological insights on the mutation rate evolution in vertebrates.


Subject(s)
Evolution, Molecular , Germ-Line Mutation , Mutation Rate , Vertebrates , Animals , Female , Male , Birds/genetics , Fishes/genetics , Germ-Line Mutation/genetics , Mammals/genetics , Reptiles/genetics , Vertebrates/genetics
2.
Nat Commun ; 13(1): 7884, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36550134

ABSTRACT

The mutation rate of a specific position in the human genome depends on the sequence context surrounding it. Modeling the mutation rate by estimating a rate for each possible k-mer, however, only works for small values of k since the data becomes too sparse for larger values of k. Here we propose a new method that solves this problem by grouping similar k-mers. We refer to the method as k-mer pattern partition and have implemented it in a software package called kmerPaPa. We use a large set of human de novo mutations to show that this new method leads to improved prediction of mutation rates and makes it possible to create models using wider sequence contexts than previous studies. As the first method of its kind, it does not only predict rates for point mutations but also insertions and deletions. We have additionally created a software package called Genovo that, given a k-mer pattern partition model, predicts the expected number of synonymous, missense, and other functional mutation types for each gene. Using this software, we show that the created mutation rate models increase the statistical power to detect genes containing disease-causing variants and to identify genes under strong selective constraint.


Subject(s)
Point Mutation , Software , Humans , Sequence Analysis, DNA/methods , Genome, Human/genetics , Mutation , Algorithms
3.
Elife ; 112022 07 27.
Article in English | MEDLINE | ID: mdl-35894300

ABSTRACT

Sequencing of cell-free DNA (cfDNA) is currently being used to detect cancer by searching both for mutational and non-mutational alterations. Recent work has shown that the length distribution of cfDNA fragments from a cancer patient can inform tumor load and type. Here, we propose non-negative matrix factorization (NMF) of fragment length distributions as a novel and completely unsupervised method for studying fragment length patterns in cfDNA. Using shallow whole-genome sequencing (sWGS) of cfDNA from a cohort of patients with metastatic castration-resistant prostate cancer (mCRPC), we demonstrate how NMF accurately infers the true tumor fragment length distribution as an NMF component - and that the sample weights of this component correlate with ctDNA levels (r=0.75). We further demonstrate how using several NMF components enables accurate cancer detection on data from various early stage cancers (AUC = 0.96). Finally, we show that NMF, when applied across genomic regions, can be used to discover fragment length signatures associated with open chromatin.


Subject(s)
Cell-Free Nucleic Acids , Circulating Tumor DNA , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Genomics/methods , Humans , Male , Mutation
5.
Elife ; 112022 01 12.
Article in English | MEDLINE | ID: mdl-35018888

ABSTRACT

In the past decade, several studies have estimated the human per-generation germline mutation rate using large pedigrees. More recently, estimates for various nonhuman species have been published. However, methodological differences among studies in detecting germline mutations and estimating mutation rates make direct comparisons difficult. Here, we describe the many different steps involved in estimating pedigree-based mutation rates, including sampling, sequencing, mapping, variant calling, filtering, and appropriately accounting for false-positive and false-negative rates. For each step, we review the different methods and parameter choices that have been used in the recent literature. Additionally, we present the results from a 'Mutationathon,' a competition organized among five research labs to compare germline mutation rate estimates for a single pedigree of rhesus macaques. We report almost a twofold variation in the final estimated rate among groups using different post-alignment processing, calling, and filtering criteria, and provide details into the sources of variation across studies. Though the difference among estimates is not statistically significant, this discrepancy emphasizes the need for standardized methods in mutation rate estimations and the difficulty in comparing rates from different studies. Finally, this work aims to provide guidelines for computational and statistical benchmarks for future studies interested in identifying germline mutations from pedigrees.


Subject(s)
Genetic Techniques , Germ-Line Mutation , Macaca mulatta/genetics , Mutation Rate , Animals , Genetic Techniques/instrumentation , Germ Cells , Laboratories , Pedigree , Reference Standards
6.
Leukemia ; 36(1): 177-188, 2022 01.
Article in English | MEDLINE | ID: mdl-34244612

ABSTRACT

Mantle cell lymphoma (MCL) is characterized by marked differences in outcome, emphasizing the need for strong prognostic biomarkers. Here, we explore expression patterns and prognostic relevance of circular RNAs (circRNAs), a group of endogenous non-coding RNA molecules, in MCL. We profiled the circRNA expression landscape using RNA-sequencing and explored the prognostic potential of 40 abundant circRNAs in samples from the Nordic MCL2 and MCL3 clinical trials, using NanoString nCounter Technology. We report a circRNA-based signature (circSCORE) developed in the training cohort MCL2 that is highly predictive of time to progression (TTP) and lymphoma-specific survival (LSS). The dismal outcome observed in the large proportion of patients assigned to the circSCORE high-risk group was confirmed in the independent validation cohort MCL3, both in terms of TTP (HR 3.0; P = 0.0004) and LSS (HR 3.6; P = 0.001). In Cox multiple regression analysis incorporating MIPI, Ki67 index, blastoid morphology and presence of TP53 mutations, circSCORE retained prognostic significance for TTP (HR 3.2; P = 0.01) and LSS (HR 4.6; P = 0.01). In conclusion, circRNAs are promising prognostic biomarkers in MCL and circSCORE improves identification of high-risk disease among younger patients treated with cytarabine-containing chemoimmunotherapy and autologous stem cell transplant.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Hematopoietic Stem Cell Transplantation/mortality , Lymphoma, Mantle-Cell/pathology , RNA, Circular/genetics , Case-Control Studies , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/therapy , Male , Middle Aged , Prognosis , RNA-Seq , Survival Rate , Transplantation, Autologous
7.
Gigascience ; 10(10)2021 10 21.
Article in English | MEDLINE | ID: mdl-34673928

ABSTRACT

The lack of consensus methods to estimate germline mutation rates from pedigrees has led to substantial differences in computational pipelines in the published literature. Here, we answer Susanne Pfeifer's opinion piece discussing the pipeline choices of our recent article estimating the germline mutation rate of rhesus macaques (Macaca mulatta). We acknowledge the differences between the method that we applied and the one preferred by Pfeifer. Yet, we advocate for full transparency and justification of choices as long as rigorous comparison of pipelines remains absent because it is the only way to conclude on best practices for the field.


Subject(s)
Germ-Line Mutation , Mutation Rate , Animals , Macaca mulatta/genetics
8.
Gigascience ; 10(5)2021 05 05.
Article in English | MEDLINE | ID: mdl-33954793

ABSTRACT

BACKGROUND: Understanding the rate and pattern of germline mutations is of fundamental importance for understanding evolutionary processes. RESULTS: Here we analyzed 19 parent-offspring trios of rhesus macaques (Macaca mulatta) at high sequencing coverage of ∼76× per individual and estimated a mean rate of 0.77 × 10-8de novo mutations per site per generation (95% CI: 0.69 × 10-8 to 0.85 × 10-8). By phasing 50% of the mutations to parental origins, we found that the mutation rate is positively correlated with the paternal age. The paternal lineage contributed a mean of 81% of the de novo mutations, with a trend of an increasing male contribution for older fathers. Approximately 3.5% of de novo mutations were shared between siblings, with no parental bias, suggesting that they arose from early development (postzygotic) stages. Finally, the divergence times between closely related primates calculated on the basis of the yearly mutation rate of rhesus macaque generally reconcile with divergence estimated with molecular clock methods, except for the Cercopithecoidea/Hominoidea molecular divergence dated at 58 Mya using our new estimate of the yearly mutation rate. CONCLUSIONS: When compared to the traditional molecular clock methods, new estimated rates from pedigree samples can provide insights into the evolution of well-studied groups such as primates.


Subject(s)
Germ-Line Mutation , Mutation Rate , Animals , Germ Cells , Macaca mulatta/genetics , Male , Phylogeny
9.
Cancer Inform ; 18: 1176935119872163, 2019.
Article in English | MEDLINE | ID: mdl-31516310

ABSTRACT

A cancer of unknown primary (CUP) is a metastatic cancer for which standard diagnostic tests fail to locate the primary cancer. As standard treatments are based on the cancer type, such cases are hard to treat and have very poor prognosis. Using molecular data from the metastatic cancer to predict the primary site can make treatment choice easier and enable targeted therapy. In this article, we first examine the ability to predict cancer type using different types of omics data. Methylation data lead to slightly better prediction than gene expression and both these are superior to classification using somatic mutations. After using 3 data types independently, we notice some differences between the classes that tend to be misclassified, suggesting that integrating the data might improve accuracy. In light of the different levels of information provided by different omics types and to be able to handle missing data, we perform multi-omics classification by hierarchically combining the classifiers. The proposed hierarchical method first classifies based on the most informative type of omics data and then uses the other types of omics data to classify samples that did not get a high confidence classification in the first step. The resulting hierarchical classifier has higher accuracy than any of the single omics classifiers and thus proves that the combination of different data types is beneficial. Our results show that using multi-omics data can improve the classification of cancer types. We confirm this by testing our method on metastatic cancers from the MET500 dataset.

10.
Methods Mol Biol ; 1910: 533-553, 2019.
Article in English | MEDLINE | ID: mdl-31278676

ABSTRACT

In this chapter, we give a short introduction to the genetics of complex diseases emphasizing evolutionary models for disease genes and the effect of different models on the genetic architecture, and we give a survey of the state-of-the-art of genome-wide association studies (GWASs).


Subject(s)
Chromosome Mapping , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Alleles , Computational Biology/methods , Confounding Factors, Epidemiologic , Evolution, Molecular , Gene Frequency , Humans , Models, Genetic , Models, Statistical
11.
Int J Cancer ; 145(12): 3445-3452, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31125115

ABSTRACT

Improved prognostic biomarkers are needed to guide personalized prostate cancer (PC) treatment decisions. Due to the prominent molecular heterogeneity of PC, multimarker panels may be more robust. Here, 25 selected top-candidate miRNA and methylation markers for PC were profiled by qPCR in malignant radical prostatectomy (RP) tissue specimens from 198 PC patients (Cohort 1, training). Using GLMnet, we trained a novel multimarker model (miMe) comprising nine miRNAs and three methylation markers that predicted postoperative biochemical recurrence (BCR) independently of the established clinicopathological CAPRA-S nomogram in Cox multivariate regression analysis in Cohort 1 (HR [95% CI]: 1.53 [1.26-1.84], p < 0.001). This result was successfully validated in two independent RP cohorts (Cohort 2, n = 159: HR [95% CI]: 1.35 [1.06-1.73], p = 0.015. TCGA, n = 350: HR [95% CI]: 1.34 [1.01-1.77], p = 0.04). Notably, in CAPRA-S low-risk patients, a high miMe score was associated with >6 times higher risk of BCR, suggesting that miMe may help identify PC patients at high risk of progression despite favorable clinicopathological factors postsurgery. Finally, miMe was a significant predictor of cancer-specific survival (p = 0.019, log-rank test) in a merged analysis of 357 RP patients. In conclusion, we trained, tested and validated a novel 12-marker panel (miMe) that showed significant independent prognostic value in three RP cohorts. In the future, combining miMe score with existing clinical nomograms may improve PC risk stratification and thus help guide treatment decisions.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Adult , Aged , Cohort Studies , Disease Progression , Humans , Kaplan-Meier Estimate , Male , Methylation , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Nomograms , Prognosis , Prostate/pathology , Prostate-Specific Antigen/genetics , Prostatectomy/methods , Prostatic Neoplasms/pathology , Risk Factors
12.
Nat Ecol Evol ; 3(5): 859, 2019 May.
Article in English | MEDLINE | ID: mdl-30988499

ABSTRACT

In the version of this article initially published, Tomas Marques-Bonet was missing the following affiliations: Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Spain; CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; and Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, Barcelona, Spain. The affiliations have been added in the PDF and HTML versions of the article.

13.
Nat Ecol Evol ; 3(2): 286-292, 2019 02.
Article in English | MEDLINE | ID: mdl-30664699

ABSTRACT

The human mutation rate per generation estimated from trio sequencing has revealed an almost linear relationship with the age of the father and the age of the mother, with fathers contributing about three times as many mutations per year as mothers. The yearly trio-based mutation rate estimate of around 0.43 × 10-9 is markedly lower than previous indirect estimates of about 1 × 10-9 per year from phylogenetic comparisons of the great apes calibrated by fossil evidence. This suggests either a slowdown in the accumulation of mutations per year in the human lineage over the past 10 million years or an inaccurate interpretation of the fossil record. Here we inferred de novo mutations in chimpanzee, gorilla, and orangutan parent-offspring trios. Extrapolating the relationship between the mutation rate and the age of parents from humans to these other great apes, we estimated that each species has higher mutation rates per year by factors of 1.50 ± 0.10, 1.51 ± 0.23, and 1.42 ± 0.22 for chimpanzee, gorilla, and orangutan, respectively, and by a factor of 1.48 ± 0.08 for the three species combined. These estimates suggest an appreciable slowdown in the yearly mutation rate in the human lineage that is likely to be recent as genome comparisons almost adhere to a molecular clock. If the nonhuman rates rather than the human rate are extrapolated over the phylogeny of the great apes, we estimate divergence and speciation times that are much more in line with the fossil record and the biogeography.


Subject(s)
Evolution, Molecular , Genetic Variation , Hominidae/genetics , Mutation , Animals , Biological Evolution , Fossils , Phylogeny
14.
Blood Adv ; 2(19): 2533-2542, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30291111

ABSTRACT

Peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS) constitutes a heterogeneous category of lymphomas, which do not fit into any of the specifically defined T-cell lymphoma entities. Both the pathogenesis and tumor biology in PTCL-NOS are poorly understood. Protein expression in pretherapeutic PTCL-NOS tumors was analyzed by proteomics. Differentially expressed proteins were compared in 3 distinct scenarios: (A) PTCL-NOS tumor tissue (n = 18) vs benign lymphoid tissue (n = 8), (B) clusters defined by principal component analysis (PCA), and (C) tumors from patients with chemosensitive vs refractory PTCL-NOS. Selected differentially expressed proteins identified by proteomics were correlated with clinico-pathological features and outcome in a larger cohort of patients with PTCL-NOS (n = 87) by immunohistochemistry (IHC). Most proteins with altered expression were identified comparing PTCL-NOS vs benign lymphoid tissue. PCA of the protein profile defined 3 distinct clusters. All benign samples clustered together, whereas PTCL-NOS tumors separated into 2 clusters with different patient overall survival rates (P = .001). Differentially expressed proteins reflected large biological diversity among PTCL-NOS, particularly associated with alterations of "immunological" pathways. The 2 PTCL-NOS subclusters defined by PCA showed disturbance of "stress-related" and "protein metabolic" pathways. α-Enolase 1 (ENO1) was found differentially expressed in all 3 analyses, and high intratumoral ENO1 expression evaluated by IHC correlated with poor outcome (hazard ratio, 2.09; 95% confidence interval, 1.17-3.73; P = .013). High expression of triosephosphate isomerase (TPI1) also showed a tendency to correlate with poor survival (P = .057). In conclusion, proteomic profiling of PTCL-NOS provided evidence of markedly altered protein expression and identified ENO1 as a novel potential prognostic marker.


Subject(s)
Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/mortality , Proteome , Proteomics , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Biomarkers, Tumor/metabolism , Chromatography, Liquid , Computational Biology , DNA-Binding Proteins/metabolism , Female , Humans , Lymphoid Tissue/metabolism , Lymphoma, T-Cell, Peripheral/genetics , Male , Phosphopyruvate Hydratase/metabolism , Prognosis , Proteomics/methods , Tandem Mass Spectrometry , Tumor Suppressor Proteins/metabolism
15.
BMC Bioinformatics ; 19(1): 147, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29673314

ABSTRACT

BACKGROUND: Detailed modelling of the neutral mutational process in cancer cells is crucial for identifying driver mutations and understanding the mutational mechanisms that act during cancer development. The neutral mutational process is very complex: whole-genome analyses have revealed that the mutation rate differs between cancer types, between patients and along the genome depending on the genetic and epigenetic context. Therefore, methods that predict the number of different types of mutations in regions or specific genomic elements must consider local genomic explanatory variables. A major drawback of most methods is the need to average the explanatory variables across the entire region or genomic element. This procedure is particularly problematic if the explanatory variable varies dramatically in the element under consideration. RESULTS: To take into account the fine scale of the explanatory variables, we model the probabilities of different types of mutations for each position in the genome by multinomial logistic regression. We analyse 505 cancer genomes from 14 different cancer types and compare the performance in predicting mutation rate for both regional based models and site-specific models. We show that for 1000 randomly selected genomic positions, the site-specific model predicts the mutation rate much better than regional based models. We use a forward selection procedure to identify the most important explanatory variables. The procedure identifies site-specific conservation (phyloP), replication timing, and expression level as the best predictors for the mutation rate. Finally, our model confirms and quantifies certain well-known mutational signatures. CONCLUSION: We find that our site-specific multinomial regression model outperforms the regional based models. The possibility of including genomic variables on different scales and patient specific variables makes it a versatile framework for studying different mutational mechanisms. Our model can serve as the neutral null model for the mutational process; regions that deviate from the null model are candidates for elements that drive cancer development.


Subject(s)
Genome, Human , Models, Genetic , Mutation Rate , Mutation/genetics , Neoplasms/genetics , Databases, Genetic , Epigenomics , Humans , Polymorphism, Single Nucleotide/genetics , Regression Analysis
16.
Blood ; 131(7): 759-770, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29208599

ABSTRACT

Mycosis fungoides (MF) is the most frequent form of cutaneous T-cell lymphoma. The disease often takes an indolent course, but in approximately one-third of the patients, the disease progresses to an aggressive malignancy with a poor prognosis. At the time of diagnosis, it is impossible to predict which patients develop severe disease and are in need of aggressive treatment. Accordingly, we investigated the prognostic potential of microRNAs (miRNAs) at the time of diagnosis in MF. Using a quantitative reverse transcription polymerase chain reaction platform, we analyzed miRNA expression in diagnostic skin biopsies from 154 Danish patients with early-stage MF. The patients were subdivided into a discovery cohort (n = 82) and an independent validation cohort (n = 72). The miRNA classifier was built using a LASSO (least absolute shrinkage and selection operator) Cox regression to predict progression-free survival (PFS). We developed a 3-miRNA classifier, based on miR-106b-5p, miR-148a-3p, and miR-338-3p, which successfully separated patients into high-risk and low-risk groups of disease progression. PFS was significantly different between these groups in both the discovery cohort and the validation cohort. The classifier was stronger than existing clinical prognostic factors and remained a strong independent prognostic tool after stratification and adjustment for these factors. Importantly, patients in the high-risk group had a significantly reduced overall survival. The 3-miRNA classifier is an effective tool to predict disease progression of early-stage MF at the time of diagnosis. The classifier adds significant prognostic value to existing clinical prognostic factors and may facilitate more individualized treatment of these patients.


Subject(s)
MicroRNAs/genetics , Mycosis Fungoides/diagnosis , Mycosis Fungoides/genetics , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Transcriptome , Biomarkers, Tumor/genetics , Denmark/epidemiology , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mycosis Fungoides/pathology , Mycosis Fungoides/therapy , Neoplasm Staging , Prognosis , Progression-Free Survival , Skin Neoplasms/pathology , Skin Neoplasms/therapy
17.
Sci Data ; 4: 170115, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28933420

ABSTRACT

Understanding of sequence diversity is the cornerstone of analysis of genetic disorders, population genetics, and evolutionary biology. Here, we present an update of our sequencing set to 15,220 Icelanders who we sequenced to an average genome-wide coverage of 34X. We identified 39,020,168 autosomal variants passing GATK filters: 31,079,378 SNPs and 7,940,790 indels. Calling de novo mutations (DNMs) is a formidable challenge given the high false positive rate in sequencing datasets relative to the mutation rate. Here we addressed this issue by using segregation of alleles in three-generation families. Using this transmission assay, we controlled the false positive rate and identified 108,778 high quality DNMs. Furthermore, we used our extended family structure and read pair tracing of DNMs to a panel of phased SNPs, to determine the parent of origin of 42,961 DNMs.


Subject(s)
Genome, Human , Humans , INDEL Mutation , Iceland , Polymorphism, Single Nucleotide
18.
Genome Res ; 27(9): 1597-1607, 2017 09.
Article in English | MEDLINE | ID: mdl-28774965

ABSTRACT

Genes in the major histocompatibility complex (MHC, also known as HLA) play a critical role in the immune response and variation within the extended 4-Mb region shows association with major risks of many diseases. Yet, deciphering the underlying causes of these associations is difficult because the MHC is the most polymorphic region of the genome with a complex linkage disequilibrium structure. Here, we reconstruct full MHC haplotypes from de novo assembled trios without relying on a reference genome and perform evolutionary analyses. We report 100 full MHC haplotypes and call a large set of structural variants in the regions for future use in imputation with GWAS data. We also present the first complete analysis of the recombination landscape in the entire region and show how balancing selection at classical genes have linked effects on the frequency of variants throughout the region.


Subject(s)
Genetic Variation/genetics , Genetics, Population , Linkage Disequilibrium/genetics , Major Histocompatibility Complex/genetics , Alleles , Chromosome Mapping , Denmark , Haplotypes/genetics , Humans , Polymorphism, Single Nucleotide/genetics
19.
J Integr Bioinform ; 14(2)2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28686574

ABSTRACT

A cancer of unknown primary (CUP) is a metastatic cancer for which standard diagnostic tests fail to identify the location of the primary tumor. CUPs account for 3-5% of cancer cases. Using molecular data to determine the location of the primary tumor in such cases can help doctors make the right treatment choice and thus improve the clinical outcome. In this paper, we present a new method for predicting the location of the primary tumor using gene expression data: locating cancers of unknown primary (LoCUP). The method models the data as a mixture of normal and tumor cells and thus allows correct classification even in impure samples, where the tumor biopsy is contaminated by a large fraction of normal cells. We find that our method provides a significant increase in classification accuracy (95.8% over 90.8%) on simulated low-purity metastatic samples and shows potential on a small dataset of real metastasis samples with known origin.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasms, Unknown Primary/genetics , Neoplasms, Unknown Primary/therapy , Biopsy , Humans
20.
PLoS One ; 11(12): e0167437, 2016.
Article in English | MEDLINE | ID: mdl-28005985

ABSTRACT

Psoriasis is a chronic cutaneous inflammatory disease. The immunopathogenesis is a complex interplay between T cells, dendritic cells and the epidermis in which T cells and dendritic cells maintain skin inflammation. Anti-tumour necrosis factor (anti-TNF)-α agents have been approved for therapeutic use across a range of inflammatory disorders including psoriasis, but the anti-inflammatory mechanisms of anti-TNF-α in lesional psoriatic skin are not fully understood. We investigated early events in skin from psoriasis patients after treatment with anti-TNF-α antibodies by use of bioinformatics tools. We used the Human Gene 1.0 ST Array to analyse gene expression in punch biopsies taken from psoriatic patients before and also 4 and 14 days after initiation of treatment with the anti-TNF-α agent adalimumab. The gene expression was analysed by gene set enrichment analysis using the Functional Annotation Tool from DAVID Bioinformatics Resources. The most enriched pathway was visualised by the Pathview Package on Kyoto Encyclopedia of Genes and Genomes (KEGG) graphs. The analysis revealed new very early events in psoriasis after adalimumab treatment. Some of these events have been described after longer periods of anti-TNF-α treatment when clinical and histological changes appear, suggesting that effects of anti-TNF-α treatment on gene expression appear very early before clinical and histological changes. Combining microarray data on biopsies from psoriasis patients with pathway analysis allowed us to integrate in vitro findings into the identification of mechanisms that may be important in vivo. Furthermore, these results may reflect primary effect of anti-TNF-α treatment in contrast to studies of gene expression changes following clinical and histological changes, which may reflect secondary changes correlated to the healing of the skin.


Subject(s)
Adalimumab/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Psoriasis/drug therapy , Skin/metabolism , Tumor Necrosis Factor-alpha/immunology , Adalimumab/pharmacology , Adult , Aged , Anti-Inflammatory Agents/pharmacology , Cytokines/genetics , Cytokines/metabolism , Gene Expression/drug effects , Hematopoietic Cell Growth Factors/genetics , Hematopoietic Cell Growth Factors/metabolism , Humans , Middle Aged , Oligonucleotide Array Sequence Analysis , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Principal Component Analysis , Psoriasis/genetics , Psoriasis/pathology , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Skin/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...