SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Lahesmaa Riitta) srt2:(2020-2024)"

Sökning: WFRF:(Lahesmaa Riitta) > (2020-2024)

  • Resultat 1-9 av 9
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Hirvonen, M Karoliina, et al. (författare)
  • Serum APOC1 levels are decreased in young autoantibody positive children who rapidly progress to type 1 diabetes
  • 2023
  • Ingår i: Scientific Reports. - : Nature Publishing Group. - 2045-2322. ; 13:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Better understanding of the early events in the development of type 1 diabetes is needed to improve prediction and monitoring of the disease progression during the substantially heterogeneous presymptomatic period of the beta cell damaging process. To address this concern, we used mass spectrometry-based proteomics to analyse longitudinal pre-onset plasma sample series from children positive for multiple islet autoantibodies who had rapidly progressed to type 1 diabetes before 4 years of age (n = 10) and compared these with similar measurements from matched children who were either positive for a single autoantibody (n = 10) or autoantibody negative (n = 10). Following statistical analysis of the longitudinal data, targeted serum proteomics was used to verify 11 proteins putatively associated with the disease development in a similar yet independent and larger cohort of children who progressed to the disease within 5 years of age (n = 31) and matched autoantibody negative children (n = 31). These data reiterated extensive age-related trends for protein levels in young children. Further, these analyses demonstrated that the serum levels of two peptides unique for apolipoprotein C1 (APOC1) were decreased after the appearance of the first islet autoantibody and remained relatively less abundant in children who progressed to type 1 diabetes, in comparison to autoantibody negative children.
  •  
2.
  • Laajala, Essi, et al. (författare)
  • Umbilical cord blood DNA methylation in children who later develop type 1 diabetes
  • 2021
  • Ingår i: European Journal of Immunology. - : John Wiley & Sons. - 0014-2980 .- 1521-4141. ; 51:Suppl. 1, s. 291-291
  • Tidskriftsartikel (övrigt vetenskapligt/konstnärligt)abstract
    • Distinct DNA methylation patterns have recently been observed to precede Type 1 Diabetes in whole blood collected from young children. Our aim was to determine if such methylation patterns are present already at the time of birth. Reduced representation bisulfite sequencing (RRBS) analysis was performed on a unique collection of umbilical cord blood samples collected within the Type 1 Diabetes Prediction and Prevention (DIPP) study. Children later diagnosed with Type 1 Diabetes and/or testing positive for multiple islet autoantibodies (N=43) were compared to control individuals (N=79), who remained autoantibody‐negative throughout the DIPP follow‐up until 15 years of age. Altogether 24 clinical and technical covariates related to the pregnancy and the mother were included in a binomial mixed effects model, which was fit separately for each high‐coverage CpG site, followed by spatial and multiple testing adjustment of P values. We discovered a strong inflation of P values, which was caused by a standard spatial adjustment method. Findings that were based on Benjamini‐Hochberg corrected spatially adjusted P values, could not be validated by Pyrosequencing. We therefore used permutation‐based significance analysis and showed that sex‐associated differentially methylated cytosines could be reproducibly detected with this approach. After empirical type 1 error control, no differences in cord blood methylation patterns were observed between cases and controls. Differences between children who progress to Type 1 Diabetes and those who remain healthy throughout childhood, are not yet present in the perinatal DNA methylome.
  •  
3.
  • Laajala, Essi, et al. (författare)
  • Umbilical cord blood DNA methylation in children who later develop type 1 diabetes
  • 2022
  • Ingår i: Diabetologia. - : Springer. - 0012-186X .- 1432-0428. ; 65:9, s. 1534-1540
  • Tidskriftsartikel (refereegranskat)abstract
    • AIMS/HYPOTHESIS: Distinct DNA methylation patterns have recently been observed to precede type 1 diabetes in whole blood collected from young children. Our aim was to determine whether perinatal DNA methylation is associated with later progression to type 1 diabetes.METHODS: Reduced representation bisulphite sequencing (RRBS) analysis was performed on umbilical cord blood samples collected within the Finnish Type 1 Diabetes Prediction and Prevention (DIPP) Study. Children later diagnosed with type 1 diabetes and/or who tested positive for multiple islet autoantibodies (n = 43) were compared with control individuals (n = 79) who remained autoantibody-negative throughout the DIPP follow-up until 15 years of age. Potential confounding factors related to the pregnancy and the mother were included in the analysis.RESULTS: No differences in the umbilical cord blood methylation patterns were observed between the cases and controls at a false discovery rate <0.05.CONCLUSIONS/INTERPRETATION: Based on our results, differences between children who progress to type 1 diabetes and those who remain healthy throughout childhood are not yet present in the perinatal DNA methylome. However, we cannot exclude the possibility that such differences would be found in a larger dataset.
  •  
4.
  • Lamichhane, Santosh, et al. (författare)
  • Circulating metabolic signatures of rapid and slow progression to type 1 diabetes in islet autoantibody-positive children
  • 2023
  • Ingår i: Frontiers in Endocrinology. - : Frontiers Media S.A.. - 1664-2392. ; 14
  • Tidskriftsartikel (refereegranskat)abstract
    • AIMS/HYPOTHESIS: Appearance of multiple islet cell autoantibodies in early life is indicative of future progression to overt type 1 diabetes, however, at varying rates. Here, we aimed to study whether distinct metabolic patterns could be identified in rapid progressors (RP, disease manifestation within 18 months after the initial seroconversion to autoantibody positivity) vs. slow progressors (SP, disease manifestation at 60 months or later from the appearance of the first autoantibody).METHODS: Longitudinal samples were collected from RP (n=25) and SP (n=41) groups at the ages of 3, 6, 12, 18, 24, or ≥ 36 months. We performed a comprehensive metabolomics study, analyzing both polar metabolites and lipids. The sample series included a total of 239 samples for lipidomics and 213 for polar metabolites.RESULTS: We observed that metabolites mediated by gut microbiome, such as those involved in tryptophan metabolism, were the main discriminators between RP and SP. The study identified specific circulating molecules and pathways, including amino acid (threonine), sugar derivatives (hexose), and quinic acid that may define rapid vs. slow progression to type 1 diabetes. However, the circulating lipidome did not appear to play a major role in differentiating between RP and SP.CONCLUSION/INTERPRETATION: Our study suggests that a distinct metabolic profile is linked with the type 1 diabetes progression. The identification of specific metabolites and pathways that differentiate RP from SP may have implications for early intervention strategies to delay the development of type 1 diabetes.
  •  
5.
  • Andrabi, Syed Bilal Ahmad, et al. (författare)
  • Long noncoding RNA LIRIL2R modulates FOXP3 levels and suppressive function of human CD4+ regulatory T cells by regulating IL2RA
  • 2024
  • Ingår i: Proceedings of the National Academy of Sciences of the United States of America. - : Proceedings of the National Academy of Sciences (PNAS). - 0027-8424 .- 1091-6490. ; 121:23
  • Tidskriftsartikel (refereegranskat)abstract
    • Regulatory T cells (Tregs) are central in controlling immune responses, and dysregulation of their function can lead to autoimmune disorders or cancer. Despite extensive studies on Tregs, the basis of epigenetic regulation of human Treg development and function is incompletely understood. Long intergenic noncoding RNAs (lincRNA)s are important for shaping and maintaining the epigenetic landscape in different cell types. In this study, we identified a gene on the chromosome 6p25.3 locus, encoding a lincRNA, that was up-regulated during early differentiation of human Tregs. The lincRNA regulated the expression of interleukin-2 receptor alpha (IL2RA), and we named it the lincRNA regulator of IL2RA (LIRIL2R). Through transcriptomics, epigenomics, and proteomics analysis of LIRIL2R-deficient Tregs, coupled with global profiling of LIRIL2R binding sites using chromatin isolation by RNA purification, followed by sequencing, we identified IL2RA as a target of LIRIL2R. This nuclear lincRNA binds upstream of the IL2RA locus and regulates its epigenetic landscape and transcription. CRISPR-mediated deletion of the LIRIL2R-bound region at the IL2RA locus resulted in reduced IL2RA expression. Notably, LIRIL2R deficiency led to reduced expression of Treg-signature genes (e.g., FOXP3, CTLA4, and PDCD1), upregulation of genes associated with effector T cells (e.g., SATB1 and GATA3), and loss of Treg-mediated suppression.
  •  
6.
  • Laajala, Essi, et al. (författare)
  • Permutation-based significance analysis reduces the type 1 error rate in bisulfite sequencing data analysis of human umbilical cord blood samples
  • 2022
  • Ingår i: Epigenetics. - : Taylor & Francis. - 1559-2294 .- 1559-2308. ; 17:12, s. 1608-1627
  • Tidskriftsartikel (refereegranskat)abstract
    • DNA methylation patterns are largely established in-utero and might mediate the impacts of in-utero conditions on later health outcomes. Associations between perinatal DNA methylation marks and pregnancy-related variables, such as maternal age and gestational weight gain, have been earlier studied with methylation microarrays, which typically cover less than 2% of human CpG sites. To detect such associations outside these regions, we chose the bisulphite sequencing approach. We collected and curated clinical data on 200 newborn infants; whose umbilical cord blood samples were analysed with the reduced representation bisulphite sequencing (RRBS) method. A generalized linear mixed-effects model was fit for each high coverage CpG site, followed by spatial and multiple testing adjustment of P values to identify differentially methylated cytosines (DMCs) and regions (DMRs) associated with clinical variables, such as maternal age, mode of delivery, and birth weight. Type 1 error rate was then evaluated with a permutation analysis. We discovered a strong inflation of spatially adjusted P values through the permutation analysis, which we then applied for empirical type 1 error control. The inflation of P values was caused by a common method for spatial adjustment and DMR detection, implemented in tools comb-p and RADMeth. Based on empirically estimated significance thresholds, very little differential methylation was associated with any of the studied clinical variables, other than sex. With this analysis workflow, the sex-associated differentially methylated regions were highly reproducible across studies, technologies, and statistical models.
  •  
7.
  • Rundquist, Olof, 1991- (författare)
  • Multi-omic time-series analysis of T-cells as a model for identification of biomarkers, treatments and upstream disease regulators
  • 2021
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • CD4+ T-cell function and their process of differentiation is a central piece of the puzzle in a multitude of diseases. CD4+ T-cells are part of the adaptive immune system and function by directing other immune cells to the site of infection and instructing B-cells to produce antibodies, among many other functions. CD4+ T-cells may differentiate into several different sub-types, such as T-helper 1, 2 and 17, with differing functions within the immune system. T-helper 1 (Th1) cells are most closely associated with the elimination of viral infections but are also associated with autoimmune diseases such as multiple sclerosis (MS) and rheumatoid arthritis (RA). T-cells develop in the thymus first as double-negative T-cells, that express neither CD4 nor CD8, going through multiple development stages before becoming double-positive T-cell that express both CD4 and CD8, before eventually giving rise to single positive CD4+ and CD8+ T-cells. This process of development is under tight control and if this control fails, cancer may result. Once CD4+ T-cells are fully developed, they may specialize as outlined above and if said process is not properly controlled, autoimmunity may result. As such, the proper understanding of these control mechanisms is of great importance for the understanding of diseases of the immune system and the discovery of biomarkers and treatments against said diseases. These control processes are often studied in a singular fashion using one omic technique, e.g., RNA sequencing (RNA-seq), with the assumption that a signal in one omic layer will be reflected in another. Recent studies attempting to integrate multiple omics have however cast doubt on this and it is becoming increasingly apparent that to gain a complete understanding of a system, the system needs to be studied at multiple levels of regulation, i.e., multiple omics.The aim of this thesis was to use multi-omics to investigate the development and differentiation process of CD4+ T-helper cells and relate it to disease mechanisms. To start, we studied T-cell development through the model of T-cell acute lymphoblastic leukaemia (T-ALL). More specifically, we studied the TET2 gene and investigated its importance in T-ALL for treatment susceptibility and mechanism in vitro. TET2 is a demethylase and functions through the removal of cytosine methylation on the DNA, a marker of gene silencing. Through treatment with decitabine, an inhibitor of DNA-methylation, and Vitamin C, a co-factor for TET2, we showed that TET2 deficient cancer cell lines were more vulnerable to treatment targeting DNA methylation and investigated the mechanistic effects of said treatment by RNA sequencing. We then moved on to study primary human naïve CD4+ T-cells and their differentiation into Th1-cells. First, we focused on T-cell activation and its importance to MS to understand the role of T-cells in mediating the lowered disease activity usually observed during pregnancy in MS. This showed that the major pregnancy hormone progesterone significantly dampens T-cell activation, providing a possible explanation for the beneficial effects of pregnancy on MS. Then, using ATAC sequencing (ATAC-seq), RNA-seq and proteomics we studied Th1-differentiation as a time series to elucidate regulatory events throughout the differentiation process and to study their implications for MS with the inclusion of progesterone treatment.  The integration of several omic techniques presents unique challenges as one does not necessarily directly translate to the other. As such, we first focused on the integration of RNA-seq and proteomics by designing a model for the prediction of protein abundance from RNA-seq and validated it through biomarker discovery. Next, we focused on the integration of ATAC-seq and RNA-seq using correlation between time series of the two techniques. This thesis provides a thorough investigation of Th1-cell differentiation and its potential involvement in disease. Time series datasets were produced to study gene regulation (ATAC-seq), gene expression (RNA-seq) and protein expression (mass spectrometry) and the work focused on their integration. This profoundly showed that through combining multiple omic techniques it was possible to gain new insights that were not possible to discover with one or the other. Multi-omic analyses are becoming more and more common in medicine today as their power to produce new insight into the complexity of complex diseases is being increasingly recognized. As such, this work forms an important foundation for future discovery of biomarkers and treatments in such diseases.
  •  
8.
  • Sen, Partho, et al. (författare)
  • Quantitative analysis of human CD4+T-cell differentiation reveals subset-specific regulation of glycosphingolipid pathways
  • 2021
  • Ingår i: European Journal of Immunology. - : John Wiley & Sons. - 0014-2980 .- 1521-4141. ; 51:Suppl. 1, s. 237-237
  • Tidskriftsartikel (övrigt vetenskapligt/konstnärligt)abstract
    • T‐cells are sentinels of adaptive immune responses. T‐cell activation, proliferation and differentiation involves metabolic reprogramming involving the interplay of genes, proteins and metabolites. Here, we aim to understand the metabolic pathways involved in the activation and functional differentiation of human CD4+ T‐cell subsets (Th1, Th2, Th17 and iTregs). We combined genome‐scale metabolic modeling, gene expression data, targeted and non‐targeted lipidomics experiments, together with in vitro gene knockdown experiments and showed that human CD4+ T cells undergo specific metabolic changes during activation and functional differentiation. In addition, we identified and confirmed the importance of ceramide and glycosphingolipid biosynthesis pathways in Th17 differentiation and effector functions. Through in vitro gene knockdown experiments, we substantiated the requirement of serine palmitoyl transferase, a de novo sphingolipid pathway in the expression of proinflammatory cytokine (IL17A and IL17F) by Th17 cells. Our findings may provide a comprehensive resource for identifying CD4+ T‐cell‐specific targets for their selective manipulation under disease conditions, particularly, diseases characterized by an imbalance of Th17/nTreg cells. 
  •  
9.
  • Sen, Partho, 1983-, et al. (författare)
  • Quantitative genome-scale metabolic modeling of human CD4+ T cell differentiation reveals subset-specific regulation of glycosphingolipid pathways
  • 2021
  • Ingår i: Cell Reports. - : Cell Press. - 2211-1247. ; 37:6
  • Tidskriftsartikel (refereegranskat)abstract
    • T cell activation, proliferation, and differentiation involve metabolic reprogramming resulting from the interplay of genes, proteins, and metabolites. Here, we aim to understand the metabolic pathways involved in the activation and functional differentiation of human CD4+ T cell subsets (T helper [Th]1, Th2, Th17, and induced regulatory T [iTreg] cells). Here, we combine genome-scale metabolic modeling, gene expression data, and targeted and non-targeted lipidomics experiments, together with in vitro gene knockdown experiments, and show that human CD4+ T cells undergo specific metabolic changes during activation and functional differentiation. In addition, we confirm the importance of ceramide and glycosphingolipid biosynthesis pathways in Th17 differentiation and effector functions. Through in vitro gene knockdown experiments, we substantiate the requirement of serine palmitoyltransferase (SPT), a de novo sphingolipid pathway in the expression of proinflammatory cytokines (interleukin [IL]-17A and IL17F) by Th17 cells. Our findings provide a comprehensive resource for selective manipulation of CD4+ T cells under disease conditions characterized by an imbalance of Th17/natural Treg (nTreg) cells.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-9 av 9

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy