Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Biochim Biophys Acta Proteins Proteom ; 1872(5): 141031, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38977230

RÉSUMÉ

Periostin is a matricellular protein known to be alternatively spliced to produce ten isoforms with a molecular weight of 78-91 kDa. Within the extracellular matrix, periostin attaches to cell surfaces to induce signaling via integrin-binding and actively participates in fibrillogenesis, orchestrating the arrangement of collagen in the extracellular environment. In atopic diseases such as atopic dermatitis (AD) and asthma, periostin is known to participate in driving the disease-causing type 2 inflammation. The periostin isoforms expressed in these diseases and the implication of the alternative splicing events are unknown. Here, we present two universal assays to map the expression of periostin isoforms at the mRNA (RT-qPCR) and protein (PRM-based mass spectrometry) levels. We use these assays to study the splicing profile of periostin in AD lesions as well as in in vitro models of AD and asthma. In these conditions, periostin displayed overexpression with isoforms 3 and 5 standing out as highly overexpressed. Notably, isoforms 9 and 10 exhibited a divergent pattern relative to the remaining isoforms. Isoforms 9 and 10 are often overlooked in periostin research and this paper presents the first evidence of their expression at the protein level. This underlines the necessity to include isoforms 9 and 10 in future research addressing periostin splice isoforms. The assays presented in this paper hold the potential to improve our insight into the splicing profile of periostin in tissues and diseases of interest. The application of these assays to AD lesions and in vitro models demonstrated their potential for identifying isoforms of particular significance, warranting a further in-depth investigation.


Sujet(s)
Épissage alternatif , Asthme , Molécules d'adhérence cellulaire , Eczéma atopique , Isoformes de protéines , Molécules d'adhérence cellulaire/génétique , Molécules d'adhérence cellulaire/métabolisme , Eczéma atopique/métabolisme , Eczéma atopique/génétique , Asthme/métabolisme , Asthme/génétique , Humains , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Spectrométrie de masse/méthodes , ARN messager/génétique , ARN messager/métabolisme ,
2.
BMJ Open ; 14(2): e077207, 2024 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-38401898

RÉSUMÉ

INTRODUCTION: Psoriasis, atopic dermatitis and contact dermatitis are common chronic inflammatory skin diseases that have a significant impact on individuals and society. METHODS AND ANALYSIS: The Copenhagen Translational Skin Immunology Biobank and Research Programme (BIOSKIN) is a translational biobank and research study that aims to prospectively collect high-quality biological samples and clinical data from 3000 patients with psoriasis, atopic dermatitis and contact dermatitis over a minimum period of 5 years. The longitudinal open design allows participants to enter and leave the study at different time points depending on their disease and treatment course. At every visit, the investigator collects biological samples, conducts interviews and assembles self-reported questionnaires on disease-specific and general health-related information. Clinical examination and biological sampling will be conducted at enrolment, during and after disease flare, before and after initiation of new treatment and at least once per year. The clinical examination includes dermatological verification of diagnosis, evaluation of disease severity and detailed information on phenotype. The biological samples include blood and when accessible and relevant, skin biopsies, tape strips and skin swabs. The data collected will undergo rigorous statistical analysis using appropriate analytical methods. As of December 2023, 825 patients have been enrolled in the study. ETHICS AND DISSEMINATION: The study is approved by the Scientific Ethical Committee of the Capital Region (H-21032986) and the Danish Data Protection Agency. Results will be published in peer-reviewed scientific journals and presented at national and international conferences.


Sujet(s)
Eczéma atopique , Eczéma de contact , Glucosamine , Psoriasis , Maladies de la peau , Humains , Eczéma atopique/thérapie , Biobanques , Maladie chronique
3.
J Invest Dermatol ; 144(7): 1608-1621.e4, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38185415

RÉSUMÉ

A substantial part of cutaneous malignant melanomas develops from benign nevi. However, the precise molecular events driving the transformation from benign to malignant melanoma are not well-understood. We used laser microdissection and mass spectrometry to analyze the proteomes of melanoma subtypes, including superficial spreading melanomas (n = 17), nodular melanomas (n = 17), and acral melanomas (n = 15). Furthermore, we compared the proteomes of nevi cells with those of melanoma cells within the same specimens (nevus-associated melanoma (n = 14)). In total, we quantified 7935 proteins. Despite the genomic and clinical differences of the melanoma subtypes, our analysis revealed relatively similar proteomes, except for the upregulation of proteins involved in immune activation in nodular melanomas versus acral melanomas. Examining nevus-associated melanoma versus nevi, we found 1725 differentially expressed proteins (false discovery rate < 0.05). Among these proteins were 140 that overlapped with cancer hallmarks, tumor suppressors, and regulators of metabolism and cell cycle. Pathway analysis indicated aberrant activation of the phosphoinositide 3-kinase-protein kinase B-mTOR pathways and the Hippo-YAP pathway. Using a classifier, we identified six proteins capable of distinguishing melanoma from nevi samples. Our study represents a comprehensive comparative analysis of the proteome in melanoma subtypes and associated nevi, offering insights into the biological behavior of these distinct entities.


Sujet(s)
Mélanome , Naevus , Protéomique , Tumeurs cutanées , Humains , Mélanome/anatomopathologie , Mélanome/métabolisme , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Protéomique/méthodes , Femelle , Naevus/anatomopathologie , Naevus/métabolisme , Mâle , Adulte d'âge moyen , Sujet âgé , Protéome/analyse , Protéome/métabolisme , Adulte , Transduction du signal , Microdissection au laser , Spectrométrie de masse ,
4.
Dermatology ; 240(1): 156-163, 2024.
Article de Anglais | MEDLINE | ID: mdl-37952520

RÉSUMÉ

BACKGROUND: Melanoma is widely recognized to be an immunogenic tumor that often contains tumor-infiltrating lymphocytes (TILs) in the tumor microenvironment. During cancer progression, expression of ligands that bind immune checkpoint (IC) proteins, such as PD-1, expressed on the surface of TILs, hinder them from exerting their antitumor functions. TILs consist of a heterogenous group of immune cells and their presence is associated with an improved overall survival in melanoma patients. Introduction of IC inhibitors has revolutionized management and prognosis of advanced melanoma. Unfortunately, the response rates have continued to be limited, resulting in growing interest in characterizing novel IC proteins, and developing combination therapy that includes inhibitors against multiple IC proteins. METHODS: In a regional cohort of 166 patients diagnosed with cutaneous superficial spreading melanoma with different degree of TILs, we investigated the tumor immune-associated gene expression profile using NanoString Technology. We used multiplex immunofluorescence (mIF) staining in a subset of tumors (N = 7), combining IC proteins T-cell immunoglobulin and ITIM domain (TIGIT) and LAG3 with a melanoma cell marker (SOX10) and immune cell markers (CD8 [cytotoxic T cells], CD4 [T helper cells], FOXP3 [regulatory T cells/Tregs], PAX5 [B cells], and CD56 [NK/NKT cells]) and IC protein PD-1. RESULTS: We found upregulation of 91 differentially expressed genes, including IC proteins, LAG3 and TIGIT in melanomas with brisk TILs compared to tumors where TILs were absent. mIF staining revealed LAG3 and TIGIT expression in the majority of CD8+ T cells. Only few Tregs and CD4+ T cells expressed LAG3, whereas majority of them expressed TIGIT. LAG3 and TIGIT were expressed in a small fraction of the NK/NKT cells and lacked in the B cells. The majority of PD-1+ cells co-localized with LAG3 and TIGIT. CONCLUSION: We report a variable expression of LAG3 and TIGIT on TILs subtypes and a coeval occurrence with PD-1. This knowledge places LAG3 and TIGIT in spatial and cellular context in melanoma. The data suggest that targeting multiple IC proteins might help overcome the current challenges with IC therapies.


Sujet(s)
Mélanome , Tumeurs cutanées , Humains , Lymphocytes T CD8+ , Lymphocytes TIL , Mélanome/anatomopathologie , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Tumeurs cutanées/anatomopathologie , Microenvironnement tumoral
5.
Dermatology ; 240(2): 317-328, 2024.
Article de Anglais | MEDLINE | ID: mdl-37935159

RÉSUMÉ

BACKGROUND: Proteins play a central role in psoriasis as they are involved in the structural phenotypic changes and inflammation that characterize the disease. This systematic review aimed to assess which proteins have been consistently reported as upregulated or downregulated in the skin and blood from patients with psoriasis. METHODS: We included proteomic studies reporting differentially expressed proteins (DEPs) in at least one of four predefined comparisons using a standardized procedure to extract and align data. Network analysis of functional protein associations was made with StringApp in Cytoscape. A protocol for this review was registered in the PROSPERO database (ref:CRD42022363226). RESULTS: We identified and assessed 772 studies published between December 2, 1996, and April 28, 2023, among which 30 studies met the inclusion and data availability criteria for analysis that together reported a sum of 5,314 DEPs. The majority of consistently reported upregulated and downregulated proteins were found in lesional versus non-lesional skin (n = 313), followed by lesional versus healthy skin (n = 185), blood from patients with psoriasis versus blood from healthy individuals (n = 140), and non-lesional versus healthy skin (n = 1). Network analysis of upregulated proteins revealed different functional clusters with interleukin (IL)-6, IL-8, IL-17A, C-C motif chemokine (CCL) 20, signal transducer and activator of transcription (STAT) 3, and interferon (IFN)-γ along with less well-studied proteins playing central roles. Some of the reported changes are associated with anti-inflammatory effects. Additionally, the proteomic dysregulation also included antimicrobial peptides, alarmins, angiogenic factors, and proteins related to protein synthesis. CONCLUSION: Our findings generally support current understandings of the pathological mechanisms in psoriasis. Importantly, some consistent findings have not been discussed before and deserve attention in future research.


Sujet(s)
Protéomique , Psoriasis , Humains , Peau/anatomopathologie , Inflammation
6.
Nucleic Acid Ther ; 33(1): 45-57, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36445751

RÉSUMÉ

Circular RNAs (circRNAs) constitute an abundant class of covalently closed noncoding RNA molecules that are formed by backsplicing from eukaryotic protein-coding genes. Recent studies have shown that circRNAs can act as microRNA or protein decoys, as well as transcriptional regulators. However, the functions of most circRNAs are still poorly understood. Because circRNA sequences overlap with their linear parent transcripts, depleting specific circRNAs without affecting host gene expression remains a challenge. In this study, we assessed the utility of LNA-modified antisense oligonucleotides (ASOs) to knock down circRNAs for loss-of-function studies. We found that, while most RNase H-dependent gapmer ASOs mediate effective knockdown of their target circRNAs, some gapmers reduce the levels of the linear parent transcript. The circRNA targeting specificity can be enhanced using design-optimized gapmer ASOs, which display potent and specific circRNA knockdown with a minimal effect on the host genes. In summary, our results demonstrate that LNA-modified ASOs complementary to backsplice-junction sequences mediate robust knockdown of circRNAs in vitro and, thus, represent a useful tool to explore the biological roles of circRNAs in loss-of-function studies in cultured cells and animal models.


Sujet(s)
Oligonucléotides antisens , ARN circulaire , Animaux , Oligonucléotides antisens/génétique , Oligonucléotides antisens/métabolisme , ARN circulaire/génétique , Oligonucléotides/génétique
7.
Sci Rep ; 12(1): 19502, 2022 11 14.
Article de Anglais | MEDLINE | ID: mdl-36376362

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with increasing incidence in western countries. Most HCC patients have advanced cancer at the time of diagnosis due to the asymptomatic nature of early-stage HCC and do not qualify for potentially curative surgical treatment, thus, highlighting the need for new therapeutic strategies. Long noncoding RNAs (lncRNAs) comprise a large and heterogeneous group of non-protein coding transcripts that play important regulatory roles in numerous biological processes in cancer. In this study, we performed RNA sequencing of liver biopsies from ten HCC, ten hepatitis C virus-associated HCC, and four normal livers to identify dysregulated lncRNAs in HCC. We show that the lncRNA p53-upregulated-regulator-of-p53-levels (PURPL) is upregulated in HCC biopsies and that its expression is p53-dependent in liver cancer cell lines. In addition, antisense oligonucleotide-mediated knockdown of PURPL inhibited cell proliferation, induced apoptosis, and sensitized HepG2 human HCC cells to treatment with the chemotherapeutic agent doxorubicin. In summary, our findings suggest that PURPL could serve as a new therapeutic target for reversing doxorubicin resistance in HCC.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , ARN long non codant , Humains , ARN long non codant/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Apoptose/génétique , Prolifération cellulaire/génétique , Doxorubicine/pharmacologie , Lignée cellulaire , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale
8.
Methods Mol Biol ; 2282: 57-75, 2021.
Article de Anglais | MEDLINE | ID: mdl-33928570

RÉSUMÉ

Small interfering RNA (siRNA)-based therapeutics holds the promise to treat a wide range of human diseases that are currently incurable using conventional therapies. Most siRNA therapeutic efforts to date have focused on the treatment of liver diseases due to major breakthroughs in the development of efficient strategies for delivering siRNA drugs to the liver. Indeed, the development of lipid nanoparticle-formulated and GalNAc-conjugated siRNA therapeutics has resulted in recent FDA approvals of the first siRNA-based drugs, patisiran for the treatment of hereditary transthyretin amyloidosis and givosiran for the treatment of acute hepatic porphyria, respectively. Here, we describe the current strategies for delivering siRNA drugs to the liver and summarize recent advances in clinical development of siRNA therapeutics for the treatment of liver diseases.


Sujet(s)
Maladies du foie/thérapie , Interférence par ARN , Petit ARN interférent/usage thérapeutique , Thérapie par l'interférence par ARN , Acétyl-galactosamine/analogues et dérivés , Acétyl-galactosamine/usage thérapeutique , Neuropathies amyloïdes familiales/génétique , Neuropathies amyloïdes familiales/métabolisme , Neuropathies amyloïdes familiales/thérapie , Animaux , Techniques de transfert de gènes , Humains , Maladies du foie/génétique , Maladies du foie/métabolisme , Porphyries hépatiques/diagnostic , Porphyries hépatiques/métabolisme , Porphyries hépatiques/thérapie , Pyrrolidines/usage thérapeutique , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme
9.
Nat Commun ; 11(1): 5587, 2020 11 05.
Article de Anglais | MEDLINE | ID: mdl-33154365

RÉSUMÉ

Human skin provides both physical integrity and immunological protection from the external environment using functionally distinct layers, cell types and extracellular matrix. Despite its central role in human health and disease, the constituent proteins of skin have not been systematically characterized. Here, we combine advanced tissue dissection methods, flow cytometry and state-of-the-art proteomics to describe a spatially-resolved quantitative proteomic atlas of human skin. We quantify 10,701 proteins as a function of their spatial location and cellular origin. The resulting protein atlas and our initial data analyses demonstrate the value of proteomics for understanding cell-type diversity within the skin. We describe the quantitative distribution of structural proteins, known and previously undescribed proteins specific to cellular subsets and those with specialized immunological functions such as cytokines and chemokines. We anticipate that this proteomic atlas of human skin will become an essential community resource for basic and translational research ( https://skin.science/ ).


Sujet(s)
Protéome/métabolisme , Peau/cytologie , Peau/métabolisme , Marqueurs biologiques/métabolisme , Cellules cultivées , Humains , Protéomique , Peau/anatomie et histologie , Peau/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE