253 results on '"KAPPA-B"'
Search Results
2. Temporal Transcript Profiling Identifies a Role for Unfolded Protein Stress in Human Gut Ischemia-Reperfusion Injury
- Author
-
Kaatje Lenaerts, Joep Grootjans, Anna M. Kip, Marco Manca, Steven W.M. Olde Damink, Bas Boonen, Cornelis H. C. Dejong, Wim A. Buurman, M'hamed Hadfoune, Joep P. M. Derikx, Erik A.L. Biessen, Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, Paediatric Surgery, ARD - Amsterdam Reproduction and Development, RS: NUTRIM - R2 - Liver and digestive health, Surgery, Pathologie, RS: Carim - B07 The vulnerable plaque: makers and markers, and MUMC+: MA Heelkunde (9)
- Subjects
ATF4, activating transcription factor 4 ,CELL-SURVIVAL ,eIF2α, eukaryotic translation initiation factor 2A ,INDUCED INFLAMMATION ,mEGF, mouse epidermal growth factor ,EM, Electron microscopy ,ROCK, Rho kinase ,Intestinal Ischemia-Reperfusion ,GADD34, growth arrest and DNA-damage-inducible protein ,Transcriptome ,ACTIVATION ,UPR, unfolded protein response ,ENDOPLASMIC-RETICULUM STRESS ,Gene expression ,I, ischemia ,Original Research ,Human Intestinal Organoids ,Gastroenterology ,DEATH ,CHOP, CCAAT/enhancer-binding protein homologous protein ,BiP, binding immunoglobulin protein ,Endoplasmic Reticulum Stress ,Cell biology ,medicine.anatomical_structure ,XBP1, X-box binding protein 1 ,Reperfusion Injury ,JNK, c-Jun N-terminal kinase ,qPCR, quantitative polymerase chain reaction ,R, reperfusion ,HYPOXIA-INDUCIBLE FACTOR ,FACTOR-I ,NF-κB, nuclear factor-κB ,IRE1, inositol-requiring enzyme 1 ,KAPPA-B ,Biology ,KEGG, Kyoto Encyclopedia of Genes and Genomes ,ER, endoplasmic reticulum ,Organoid ,medicine ,KINASE ,Integrated stress response ,Humans ,C, control ,Transcriptomics ,GO, gene ontology ,Hepatology ,Microarray analysis techniques ,XBP1s, spliced X-box binding protein 1 ,Endoplasmic reticulum ,PERK, protein kinase R-like ER kinase ,Activating Transcription Factor 4 ,Small intestine ,ISRIB, integrated stress response inhibitor ,ER ,Unfolded protein response ,Unfolded Protein Response ,HIF1A, hypoxia-inducible factor 1-α ,Transcription Factor CHOP ,MAPK, mitogen-activated protein kinase - Abstract
Background & Aims Intestinal ischemia-reperfusion injury is a serious and life-threatening condition. A better understanding of molecular mechanisms related to intestinal ischemia-reperfusion injury in human beings is imperative to find therapeutic targets and improve patient outcome. Methods First, the in vivo dynamic modulation of mucosal gene expression of the ischemia-reperfusion–injured human small intestine was studied. Based on functional enrichment analysis of the changing transcriptome, one of the predominantly regulated pathways was selected for further investigation in an in vitro human intestinal organoid model. Results Ischemia-reperfusion massively changed the transcriptional landscape of the human small intestine. Functional enrichment analysis based on gene ontology and pathways pointed to the response to unfolded protein as a predominantly regulated process. In addition, regulatory network analysis identified hypoxia-inducing factor 1A as one of the key mediators of ischemia-reperfusion–induced changes, including the unfolded protein response (UPR). Differential expression of genes involved in the UPR was confirmed using quantitative polymerase chain reaction analysis. Electron microscopy showed signs of endoplasmic reticulum stress. Collectively, these findings point to a critical role for unfolded protein stress in intestinal ischemia-reperfusion injury in human beings. In a human intestinal organoid model exposed to hypoxia-reoxygenation, attenuation of UPR activation with integrated stress response inhibitor strongly reduced pro-apoptotic activating transcription factor 4 (ATF4)-CCAAT/enhancer-binding protein homologous protein (CHOP) signaling. Conclusions Transcriptome analysis showed a crucial role for unfolded protein stress in the response to ischemia-reperfusion in human small intestine. UPR inhibition during hypoxia-reoxygenation in an intestinal organoid model suggests that downstream protein kinase R-like ER kinase (PERK) signaling may be a promising target to reduce intestinal ischemia-reperfusion injury. Microarray data are available in GEO (https://www.ncbi.nlm.nih.gov/gds, accession number GSE37013)., Graphical abstract
- Published
- 2022
3. MANF regulates neuronal survival and UPR through its ER-located receptor IRE1a
- Author
-
Vera Kovaleva, Li-Ying Yu, Larisa Ivanova, Olesya Shpironok, Jinhan Nam, Ave Eesmaa, Esa-Pekka Kumpula, Sven Sakson, Urve Toots, Mart Ustav, Juha T. Huiskonen, Merja H. Voutilainen, Päivi Lindholm, Mati Karelson, Mart Saarma, Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, Division of Pharmacology and Pharmacotherapy, Regenerative Neuroscience, Laboratory of Structural Biology, Molecular and Integrative Biosciences Research Programme, Helsinki Institute of Sustainability Science (HELSUS), Drug Research Program, Divisions of Faculty of Pharmacy, and Mart Saarma / Principal Investigator
- Subjects
Unfolded protein response ,Transmembrane protein ,Endoplasmic-reticulum stress ,Proliferation ,3112 Neurosciences ,Neurotrophic factor ,Beta ,Activation ,Homeostasis ,Kappa-b ,General Biochemistry, Genetics and Molecular Biology ,Model - Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-located pro-tein with cytoprotective effects in neurons and pancreatic b cells in vitro and in models of neurodegeneration and diabetes in vivo. However, the exact mode of MANF action has remained elusive. Here, we show that MANF directly interacts with the ER transmembrane unfolded protein response (UPR) sensor IRE1a, and we identify the binding interface between MANF and IRE1a. The expression of wild-type MANF, but not its IRE1a binding-deficient mutant, attenuates UPR signaling by decreasing IRE1a oligomerization; phosphor-ylation; splicing of Xbp1, Atf6, and Txnip levels; and protecting neurons from ER stress-induced death. MANF-IRE1a interaction and not MANF-BiP interaction is crucial for MANF pro-survival activity in neurons in vitro and is required to protect dopamine neurons in an animal model of Parkinson's disease. Our data show IRE1a as an intracellular receptor for MANF and regulator of neuronal survival.
- Published
- 2023
4. Spirulina, wakame or goji berries do not lower markers of low-grade systemic inflammation in healthy subjects
- Author
-
Jose J. Van den Driessche, Ronald P. Mensink, Jogchum Plat, RS: NUTRIM - R1 - Obesity, diabetes and cardiovascular health, and Nutrition and Movement Sciences
- Subjects
human intervention study ,INTERLEUKIN-8 ,Nutrition and Dietetics ,CYTOKINES ,INHIBITION ,Medicine (miscellaneous) ,low-grade systemic inflammation ,EXTRACT ,goji berries ,KAPPA-B ,Biochemistry ,wakame ,DOUBLE-BLIND ,POLYSACCHARIDE-PROTEIN COMPLEX ,EPIDEMIOLOGY ,spirulina ,SPOROPHYLL ,OXIDATIVE STRESS ,Food Science - Abstract
Introduction: We have earlier reported that consumption of the algae spirulina (Arthrospira platensis or maxima) and wakame (Undaria pinnatifida) for 17 days, and a single dose of goji berries (Lycium barbarum) did not affect fasting or postprandial CVD risk markers in healthy subjects. However, evidence is increasing that low-grade systemic inflammation is also an important marker for CVD risk. Based on information from in vitro and animal studies, we hypothesize that both consumption of the algae spirulina and wakame as well as a single dose of goji berries lowers markers for low-grade inflammation. Methods: Two randomized, placebo-controlled, crossover trials were performed. In the algae study, 35 non-hypercholesterolemic, healthy subjects consumed 4.8 grams of spirulina, wakame or placebo for 17 days, separated by 14-day washout periods. After 17 days, fasting serum TNFα, IL-6, IL-8, and hsCRP concentrations were measured. In the goji berry study, 17 healthy, overweight men received a mixed meal with or without 25 grams of dried goji berries. Before and up to 4 hours after meal intake, serum concentrations of TNFα, IL-6 and IL-8 were measured. Results: Consumption of spirulina or wakame did not affect serum concentrations of TNFα, IL-6, IL-8 or hsCRP. In the goji berry study, serum IL-6 and IL-8 concentrations increased postprandially. For IL-8, these increases were more pronounced after the goji berry meal compared to the control meal (P = 0.003). No effects on TNFα were observed. Conclusion: 17 days of spirulina or wakame consumption, or a single dose of goji berries did not lower markers of low-grade systemic inflammation in healthy, non-immunocompromised subjects. It is plausible that anti-inflammatory effects of these interventions can only be expected in subjects with an inflammatory risk.Keywords: spirulina, wakame, goji berries, low-grade systemic inflammation, cytokines
- Published
- 2021
5. Potential roles of super enhancers in inflammatory gene transcription
- Author
-
Higashijima, Yoshiki, Kanki, Yasuharu, Higashijima, Yoshiki, and Kanki, Yasuharu
- Abstract
Acute and chronic inflammation is a basic pathological event that contributes to atherosclerosis, cancer, infectious diseases, and immune disorders. Inflammation is an adaptive process to both external and internal stimuli experienced by the human body. Although the mechanism of gene transcription is highly complicated and orchestrated in a timely and spatial manner, recent developments in next-generation sequencing, genome-editing, cryo-electron microscopy, and single cell-based technologies could provide us with insights into the roles of super enhancers (SEs). Initially, SEs were implicated in determining cell fate; subsequent studies have clarified that SEs are associated with various pathological conditions, including cancer and inflammatory diseases. Recent technological advances have unveiled the molecular mechanisms of SEs, which involve epigenetic histone modifications, chromatin three-dimensional structures, and phase-separated condensates. In this review, we discuss the relationship between inflammation and SEs and the therapeutic potential of SEs for inflammatory diseases.
- Published
- 2022
6. Detection of Neanderthal Adaptively Introgressed Genetic Variants That Modulate Reporter Gene Expression in Human Immune Cells
- Author
-
Jagoda, Evelyn, Xue, James R., Reilly, Steven K., Dannemann, Michael, Racimo, Fernando, Huerta-Sanchez, Emilia, Sankararaman, Sriram, Kelso, Janet, Pagani, Luca, Sabeti, Pardis C., Capellini, Terence D., Jagoda, Evelyn, Xue, James R., Reilly, Steven K., Dannemann, Michael, Racimo, Fernando, Huerta-Sanchez, Emilia, Sankararaman, Sriram, Kelso, Janet, Pagani, Luca, Sabeti, Pardis C., and Capellini, Terence D.
- Abstract
Although some variation introgressed from Neanderthals has undergone selective sweeps, little is known about its functional significance. We used a Massively Parallel Reporter Assay (MPRA) to assay 5,353 high-frequency introgressed variants for their ability to modulate the gene expression within 170 bp of endogenous sequence. We identified 2,548 variants in active putative cis-regulatory elements (CREs) and 292 expression-modulating variants (emVars). These emVars are predicted to alter the bindingmotifs of important immune transcription factors, are enriched for associations with neutrophil and white blood cell count, and are associated with the expression of genes that function in innate immune pathways including inflammatory response and antiviral defense. We combined theMPRA data with other data sets to identify strong candidates to be driver variants of positive selection including an emVar that may contribute to protection against severe COVID-19 response. We endogenously deleted two CREs containing expression-modulation variants linked to immune function, rs11624425 and rs80317430, identifying their primary genic targets as ELMSAN1, and PAN2 and STAT2, respectively, three genes differentially expressed during influenza infection. Overall, we present the first database of experimentally identified expression-modulating Neanderthal-introgressed alleles contributing to potential immune response in modern humans.
- Published
- 2022
7. Immunoinflammatory, Thrombohaemostatic, and Cardiovascular Mechanisms in COVID-19
- Subjects
NITRIC-OXIDE ,SARS-CoV-2 ,BLOCKADE ,INHIBITION ,MYOCARDITIS ,COVID-19 ,ACE2 ,KAPPA-B ,CORONAVIRUS ,SARS-COV-2 RECEPTOR ACE2 ,ANGIOTENSIN-ALDOSTERONE SYSTEM ,RAAS ,ADIPOSE-TISSUE ,ATHEROSCLEROSIS ,cardiovascular disease ,inflammation ,thrombosis - Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has deranged the recent history of humankind, afflicting more than 27 million individuals to date. While the majority of COVID-19 patients recuperate, a considerable number of patients develop severe complications. Bilateral pneumonia constitutes the hallmark of severe COVID-19 disease but an involvement of other organ systems, namely the cardiovascular system, kidneys, liver, and central nervous system, occurs in at least half of the fatal COVID-19 cases. Besides respiratory failure requiring ventilation, patients with severe COVID-19 often display manifestations of systemic inflammation and thrombosis as well as diffuse microvascular injury observed postmortem. In this review, we survey the mechanisms that may explain how viral entry and activation of endothelial cells by severe acute respiratory syndrome coronavirus 2 can give rise to a series of events including systemic inflammation, thrombosis, and microvascular dysfunction. This pathophysiological scenario may be particularly harmful in patients with overt cardiovascular disease and may drive the fatal aspects of COVID-19. We further shed light on the role of the renin-angiotensin aldosterone system and its inhibitors in the context of COVID-19 and discuss the potential impact of antiviral and anti-inflammatory treatment options. Acknowledging the comorbidities and potential organ injuries throughout the course of severe COVID-19 is crucial in the clinical management of patients affecting treatment approaches and recovery rate.
- Published
- 2020
8. Immunoinflammatory, Thrombohaemostatic, and Cardiovascular Mechanisms in COVID-19
- Author
-
Christian Weber, Selin Gencer, Emiel P. C. van der Vorst, Yvonne Döring, Michael Lacy, and Dorothee Atzler
- Subjects
0301 basic medicine ,medicine.medical_specialty ,BLOCKADE ,Central nervous system ,INHIBITION ,MYOCARDITIS ,ACE2 ,Context (language use) ,KAPPA-B ,CORONAVIRUS ,Disease ,030204 cardiovascular system & hematology ,Systemic inflammation ,SARS-COV-2 RECEPTOR ACE2 ,ANGIOTENSIN-ALDOSTERONE SYSTEM ,Renin-Angiotensin System ,03 medical and health sciences ,0302 clinical medicine ,RAAS ,cardiovascular disease ,medicine ,Humans ,Intensive care medicine ,Pandemics ,Inflammation ,Hemostasis ,NITRIC-OXIDE ,business.industry ,SARS-CoV-2 ,Microcirculation ,Immunity ,COVID-19 ,Thrombosis ,Hematology ,medicine.disease ,Theme Issue Article ,Pathophysiology ,3. Good health ,ADIPOSE-TISSUE ,030104 developmental biology ,medicine.anatomical_structure ,ATHEROSCLEROSIS ,Respiratory failure ,Cardiovascular Diseases ,medicine.symptom ,business - Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has deranged the recent history of humankind, afflicting more than 27 million individuals to date. While the majority of COVID-19 patients recuperate, a considerable number of patients develop severe complications. Bilateral pneumonia constitutes the hallmark of severe COVID-19 disease but an involvement of other organ systems, namely the cardiovascular system, kidneys, liver, and central nervous system, occurs in at least half of the fatal COVID-19 cases. Besides respiratory failure requiring ventilation, patients with severe COVID-19 often display manifestations of systemic inflammation and thrombosis as well as diffuse microvascular injury observed postmortem. In this review, we survey the mechanisms that may explain how viral entry and activation of endothelial cells by severe acute respiratory syndrome coronavirus 2 can give rise to a series of events including systemic inflammation, thrombosis, and microvascular dysfunction. This pathophysiological scenario may be particularly harmful in patients with overt cardiovascular disease and may drive the fatal aspects of COVID-19. We further shed light on the role of the renin–angiotensin aldosterone system and its inhibitors in the context of COVID-19 and discuss the potential impact of antiviral and anti-inflammatory treatment options. Acknowledging the comorbidities and potential organ injuries throughout the course of severe COVID-19 is crucial in the clinical management of patients affecting treatment approaches and recovery rate.
- Published
- 2020
9. Early vascular ageing in chronic kidney disease
- Subjects
cardiovascular ,CELLULAR SENESCENCE ,KAPPA-B ,CALCIFICATION ,NRF2 ,SUPPLEMENTATION ,MECHANISMS ,ACTIVATION ,PATHWAY ,age ,DNA-DAMAGE ,inflammation ,CKD ,oxidative stress - Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by cardiovascular disease, persistent uraemic inflammation, osteoporosis muscle wasting and frailty. The accelerated early vascular ageing (EVA) process mediated by medial vascular calcification (VC) is a hallmark of senescence as well as a strong predictor of cardiovascular morbidity and mortality in the CKD population. Current clinical therapeutic strategies and novel treatments for VC have not yet been proven to prevent or reverse VC progression in patients with CKD. Knowledge of the fundamental mechanism underlying EVA is urgently needed to identify and develop novel and efficient therapeutic targets for VC and EVA. An accumulating body of evidence indicates that deoxyribonucleic acid (DNA) damage-induced cellular senescence and 'inflammaging' may largely contribute to such pathological conditions characterized by accelerated EVA. Growing evidence shows that nuclear factor erythroid 2-related factor 2 (NRF2) signalling and vitamin K play a crucial role in counteracting oxidative stress, DNA damage, senescence and inflammaging, whereby NRF2 activation and vitamin K supplementation may provide a novel treatment target for EVA. In this review we discuss the link between senescence and EVA in the context of CKD, with a focus on the role of NRF2 and vitamin K in DNA damage signalling, senescence and inflammaging.
- Published
- 2020
10. Early vascular ageing in chronic kidney disease: impact of inflammation, vitamin K, senescence and genomic damage
- Author
-
Peter Stenvinkel, Paul G. Shiels, Leon J. Schurgers, and Lu Dai
- Subjects
0301 basic medicine ,Senescence ,Vitamin K ,DNA damage ,NF-E2-Related Factor 2 ,Population ,Reviews ,Inflammation ,Context (language use) ,KAPPA-B ,030204 cardiovascular system & hematology ,Bioinformatics ,medicine.disease_cause ,NRF2 ,SUPPLEMENTATION ,MECHANISMS ,ACTIVATION ,PATHWAY ,03 medical and health sciences ,0302 clinical medicine ,medicine ,CKD ,Humans ,Renal Insufficiency, Chronic ,education ,Vascular Calcification ,Cellular Senescence ,Transplantation ,education.field_of_study ,business.industry ,cardiovascular ,medicine.disease ,CALCIFICATION ,Oxidative Stress ,030104 developmental biology ,DNA-DAMAGE ,age ,Nephrology ,Cardiovascular Diseases ,Disease Progression ,medicine.symptom ,business ,Cell aging ,Oxidative stress ,Kidney disease ,DNA Damage - Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by cardiovascular disease, persistent uraemic inflammation, osteoporosis muscle wasting and frailty. The accelerated early vascular ageing (EVA) process mediated by medial vascular calcification (VC) is a hallmark of senescence as well as a strong predictor of cardiovascular morbidity and mortality in the CKD population. Current clinical therapeutic strategies and novel treatments for VC have not yet been proven to prevent or reverse VC progression in patients with CKD. Knowledge of the fundamental mechanism underlying EVA is urgently needed to identify and develop novel and efficient therapeutic targets for VC and EVA. An accumulating body of evidence indicates that deoxyribonucleic acid (DNA) damage–induced cellular senescence and ‘inflammaging’ may largely contribute to such pathological conditions characterized by accelerated EVA. Growing evidence shows that nuclear factor erythroid 2–related factor 2 (NRF2) signalling and vitamin K play a crucial role in counteracting oxidative stress, DNA damage, senescence and inflammaging, whereby NRF2 activation and vitamin K supplementation may provide a novel treatment target for EVA. In this review we discuss the link between senescence and EVA in the context of CKD, with a focus on the role of NRF2 and vitamin K in DNA damage signalling, senescence and inflammaging.
- Published
- 2020
11. Proteasome Inhibitor Immunotherapy for the Epithelial to Mesenchymal Transition: Assessing the A549 Lung Cancer Cell Microenvironment and the Role of M1, M2a and M2c ‘Hydrocortisone-Polarised’ Macrophages
- Author
-
Selin Engür-Öztürk and Miriş Dikmen
- Subjects
Epithelial-Mesenchymal Transition ,Lung Neoplasms ,Hydrocortisone ,Macrophages ,Emt ,Mln2238 ,General Medicine ,Marker ,respiratory system ,Ixazomib ,Metastasis ,Bortezomib ,A549 ,Tumor Microenvironment ,Genetics ,Alternative Activation ,Humans ,NF-kB ,Tumor-Growth ,Immunotherapy ,Kappa-B ,Proteasome Inhibitors ,Molecular Biology - Abstract
Lung cancer is a leading cause of cancer-related deaths, primarily as a result of metastases. In this metastasis, the epithelial-to-mesenchymal transition (EMT) is essential. Interaction with the cancer cell microenvironment is primarily dependent on M1- and M2-polarized macrophage. In this study, we revealed the EMT-associated activity of M1, M2a and M2c macrophages in A549 lung cancer cells. We established a co-culture model of A549 lung cancer cells utilizing THP-1-derived M1/M2 polarised macrophages to explore the involvement of macrophages in the immune response, apoptosis, and EMT in lung cancer. Although multiple polarising agents are routinely used for M1 and M2 conversion, we assessed a new possible polarising agent, hydrocortisone. M1 increased A549 cell sensitivity to proteasome inhibitors and decreased A549 cell viability by inducing apoptosis. EMT was induced in the presence of M2c macrophages in A549 cells by the levels of vimentin, fibronectin, E-cadherin, NF-kB, CCL-17. We also revealed the antiproliferative effects of bortezomib and ixazomib on A549 cells in both 2D and 3D cultures. Our findings could help develop an immunotherapeutic strategy by shedding light on a previously undiscovered part of the EMT pathway. Furthermore, additional investigation may reveal that polarising tumour-associated macrophages to M1 and eliminating the M2a or particularly the M2c subtype are effective anti-cancer strategies.
- Published
- 2022
12. Detection of Neanderthal adaptively introgressed genetic variants that modulate reporter gene expression in human immune cells
- Author
-
Evelyn Jagoda, Pardis C. Sabeti, Michael Dannemann, James R. Xue, Steven K. Reilly, Fernando Racimo, Sriram Sankararaman, Terence D. Capellini, Emilia Huerta-Sanchez, Luca Pagani, and Janet Kelso
- Subjects
SELECTION ,introgression ,Gene Expression ,adaptation ,NUCLEAR-FACTOR ,KAPPA-B ,Biology ,AcademicSubjects/SCI01180 ,SCREEN REVEALS ,LINKS ,massively parallel reporter assay ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,positive selection ,Gene expression ,Genetics ,Animals ,Humans ,TRANSCRIPTION ,Allele ,Molecular Biology ,Transcription factor ,Gene ,Discoveries ,Ecology, Evolution, Behavior and Systematics ,Neanderthals ,030304 developmental biology ,Inflammation ,0303 health sciences ,Reporter gene ,Innate immune system ,LANDSCAPE ,Genome, Human ,AcademicSubjects/SCI01130 ,Genetic Variation ,neandertal ,Immunity, Innate ,GENOME ,ADMIXTURE ,immune ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Although some variation introgressed from Neanderthals has undergone selective sweeps, little is known about its functional significance. We used a Massively Parallel Reporter Assay (MPRA) to assay 5,353 high-frequency introgressed variants for their ability to modulate the gene expression within 170 bp of endogenous sequence. We identified 2,548 variants in active putative cis-regulatory elements (CREs) and 292 expression-modulating variants (emVars). These emVars are predicted to alter the binding motifs of important immune transcription factors, are enriched for associations with neutrophil and white blood cell count, and are associated with the expression of genes that function in innate immune pathways including inflammatory response and antiviral defense. We combined the MPRA data with other data sets to identify strong candidates to be driver variants of positive selection including an emVar that may contribute to protection against severe COVID-19 response. We endogenously deleted two CREs containing expression-modulation variants linked to immune function, rs11624425 and rs80317430, identifying their primary genic targets as ELMSAN1, and PAN2 and STAT2, respectively, three genes differentially expressed during influenza infection. Overall, we present the first database of experimentally identified expression-modulating Neanderthal-introgressed alleles contributing to potential immune response in modern humans.
- Published
- 2022
13. The arginine methyltransferase PRMT7 promotes extravasation of monocytes resulting in tissue injury in COPD
- Author
-
Gizem Günes Günsel, Thomas M. Conlon, Aicha Jeridi, Rinho Kim, Zeynep Ertüz, Niklas J. Lang, Meshal Ansari, Mariia Novikova, Dongsheng Jiang, Maximilian Strunz, Mariia Gaianova, Christine Hollauer, Christina Gabriel, Ilias Angelidis, Sebastian Doll, Jeanine C. Pestoni, Stephanie L. Edelmann, Marlene Sophia Kohlhepp, Adrien Guillot, Kevin Bassler, Hannelore P. Van Eeckhoutte, Özgecan Kayalar, Nur Konyalilar, Tamara Kanashova, Sophie Rodius, Carolina Ballester-López, Carlos M. Genes Robles, Natalia Smirnova, Markus Rehberg, Charu Agarwal, Ioanna Krikki, Benoit Piavaux, Stijn E. Verleden, Bart Vanaudenaerde, Melanie Königshoff, Gunnar Dittmar, Ken R. Bracke, Joachim L. Schultze, Henrik Watz, Oliver Eickelberg, Tobias Stoeger, Gerald Burgstaller, Frank Tacke, Vigo Heissmeyer, Yuval Rinkevich, Hasan Bayram, Herbert B. Schiller, Marcus Conrad, Robert Schneider, Ali Önder Yildirim, Kayalar, Özgecan, Konyalılar, Nur, Bayram, Hasan (ORCID 0000-0002-5236-766X & YÖK ID 4890), Yıldırım, Ali Önder, Günsel, Gizem Güneş, Conlon, Thomas M., Jeridi, Aicha, Kim, Rinho, Ertuez, Zeynep, Lang, Niklas J., Ansari, Meshal, Novikova, Mariia, Jiang, Dongsheng, Strunz, Maximilian, Gaianova, Mariia, Hollauer, Christine, Gabriel, Christina, Angelidis, Ilias, Doll, Sebastian, Pestoni, Jeanine C., Edelmann, Stephanie L., Kohlhepp, Marlene Sophia, Guillot, Adrien, Bassler, Kevin, Van Eeckhoutte, Hannelore P., Kanashova, Tamara, Rodius, Sophie, Ballester-Lopez, Carolina, Robles, Carlos M. Genes, Smirnova, Natalia, Rehberg, Markus, Agarwal, Charu, Krikki, Ioanna, Piavaux, Benoit, Verleden, Stijn E., Vanaudenaerde, Bart, Koenigshoff, Melanie, Dittmar, Gunnar, Bracke, Ken R., Schultze, Joachim L., Watz, Henrik, Eickelberg, Oliver, Stoeger, Tobias, Burgstaller, Gerald, Tacke, Frank, Heissmeyer, Vigo, Rinkevich, Yuval, Schiller, Herbert B., Conrad, Marcus, Schneider, Robert, Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), and School of Medicine
- Subjects
EXPRESSION ,Protein-Arginine N-Methyltransferases ,metabolism [Histones] ,metabolism [Arginine] ,PRMT7 protein, human ,General Physics and Astronomy ,KAPPA-B ,PRMT2 protein, human ,Arginine ,Animals ,Histones ,Humans ,Intracellular signaling peptides and proteins ,Mice ,Monocytes ,Protein-arginine n-methyltransferases ,Pulmonary disease ,Chronic obstructive ,General Biochemistry, Genetics and Molecular Biology ,Multidisciplinary sciences ,Pulmonary Disease, Chronic Obstructive ,INFLAMMATION ,PRMT7 protein, mouse ,SMOKE-INDUCED EMPHYSEMA ,Medicine and Health Sciences ,PHOSPHORYLATION ,metabolism [Protein-Arginine N-Methyltransferases] ,RAP1 ,Multidisciplinary ,METHYLATION ,Intracellular Signaling Peptides and Proteins ,Biology and Life Sciences ,General Chemistry ,DIFFERENTIATION ,CELLS ,genetics [Pulmonary Disease, Chronic Obstructive] ,ddc:500 ,Human medicine ,Technology Platforms ,ELASTASE ,metabolism [Monocytes] - Abstract
Extravasation of monocytes into tissue and to the site of injury is a fundamental immunological process, which requires rapid responses via post translational modifications (PTM) of proteins. Protein arginine methyltransferase 7 (PRMT7) is an epigenetic factor that has the capacity to mono-methylate histones on arginine residues. Here we show that in chronic obstructive pulmonary disease (COPD) patients, PRMT7 expression is elevated in the lung tissue and localized to the macrophages. In mouse models of COPD, lung fibrosis and skin injury, reduced expression of PRMT7 associates with decreased recruitment of monocytes to the site of injury and hence less severe symptoms. Mechanistically, activation of NF-kappa B/RelA in monocytes induces PRMT7 transcription and consequential mono-methylation of histones at the regulatory elements of RAP1A, which leads to increased transcription of this gene that is responsible for adhesion and migration of monocytes. Persistent monocyte-derived macrophage accumulation leads to ALOX5 over-expression and accumulation of its metabolite LTB4, which triggers expression of ACSL4 a ferroptosis promoting gene in lung epithelial cells. Conclusively, inhibition of arginine mono-methylation might offer targeted intervention in monocyte-driven inflammatory conditions that lead to extensive tissue damage if left untreated. Chronic obstructive pulmonary disease is a progressive and incurable chronic condition that involves accumulation of inflammatory macrophages in the lung tissue. Authors here show in mouse models of lung disease that PRMT7, a protein arginine methyltransferase, is an important regulator of recruitment and the pro-inflammatory phenotype of macrophages., German Center for Lung Research (DZL); German Research Foundation; Deutsche Forschungsgemeinschaft (DFG); DFG Priority Program 2306; German Bundesministerium für Bildung und Forschung (BMBF) VIP + program NeuroProtect; Ministry of Science and Higher Education of the Russian Federation; Else Kröner-Fresenius-Stiftung; European Union (EU); Horizon 2020; European Research Council (ERC); Research and Innovation Program; Projekt DEAL
- Published
- 2022
14. Clinical and molecular evaluation of MEFV gene variants in the Turkish population: a study by the National Genetics Consortium
- Author
-
ERTEN, ŞÜKRAN, DÜNDAR, MUNİS, Altinay, Mert, Bakir-Gungor, Burcu, TEMEL, ŞEHİME GÜLSÜN, AKIN, HALUK, ARTAN, SEVİLHAN, Acar, Aynur, Cora, Tulin, ŞAHİN, FERİDE İFFET, DURSUN, AHMET, Sezer, Ozlem, GÜRKAN, HAKAN, Erdogan, Murat, Kebudi, Rejin, ÇİLİNGİR, OĞUZ, AYKUT, AYÇA, Durmaz, Burak, EMMUNGİL, HAKAN, KARACA, EMİN, Emekli, Rabia, Gonen, Gizem Akinci, Onay, Huseyin, DURMAZ, ASUDE, Balta, Burhan, Aynekin, Busra, KANDEMİR, NEFİSE, Kiraz, Aslihan, ÇOĞULU, MUHSİN ÖZGÜR, Gunes, Meltem Cerrah, KARADUMAN, NESLİHAN, Ozkayin, Nese, ÖZKINAY, FERİŞTAH FERDA, YALÇINTEPE, SİNEM, ÇOLAK, Fatma, SUBAŞIOĞLU, Aslı, Haziyeva, Konul, Bayramicli, Oya Uygur, Bilge, Ilmay, Kaya, Niyazi, Bayram, Arslan, Erguzeloglu, Cemre Ornek, KAVUKÇU, SALİH, DOĞAN, BERKCAN, Tuncel, Gulten, Mocan, Gamze, Kale, Hamdi, Gurakan, Figen, Uyguner, Zehra Oya, Tunc, Betul, Kuru, Seda, Boz, Mehmet, Dundar, Ayca, AKALIN, HİLAL, KAZIMLI, ULVIYYA, Zeybel, Mujdat, BAYSAL, KÜBRA, Zamani, Aysegul, GEÇKİNLİ, BİLGEN BİLGE, Uzel, Veysiye Hulya, DURAK ARAS, BEYHAN, Kiranatlioglu, Kubra, Ates, Esra Arslan, KULAK ABAY, HANDE, COŞKUN, MERT, EM, SERDA, ALTIOK CLARK, ÖZDEN, TOYLU, ASLI, TOZKIR, HİLMİ, Komesli, Zeynep, KOCAGİL, SİNEM, ÇEVİK, MUHAMMER ÖZGÜR, Eroz, Recep, Demirtas, Mercan, FIRAT, CEM KORAY, ERGÜN, MEHMET ALİ, YÜCE KAHRAMAN, Çiğdem, Yigit, Serbulent, Sanri, Aslihan, Siniksaran, Betul Seyhan, DEMİR, MİKAİL, ÖZÇELİK, FIRAT, Dundar, Bilge, BAŞ, HASAN, SUSAM, EZGİ, Karakoyun, Hilal Keskin, KARASU, NİLGÜN, Kenanoglu, Sercan, SAATÇİ, ÇETİN, ÖZKUL, YUSUF, Temena, Arda, Yuksel, Berrin, ÇAĞLAYAN, AHMET OKAY, BAHADIR, Oğuzhan, Genc, Gunes Cakmak, KEKLİKCİ, ALİ RIZA, Altunoglu, Umut, Sarac, Elif, Baskin, Esra Sidika, TOSUN, ÖZGÜR, Tulay, Pinar, Kabayegit, Zehra Manav, Altan, Mustafa, Mardan, Lamiya, Sayar, Ceyhan, ERZURUMLUOĞLU GÖKALP, EBRU, ÇETİN, GÖKHAN OZAN, Turkgenc, Burcu, Arslan, Serap, Tumer, Sait, NUR, BANU, Ergoren, Mahmut Cerkez, Onder, Nerin Bahceciler, KOÇAK, NADİR, Tasdemir, Mehmet, NERGİZ, SÜLEYMAN, Beyitler, Ilke, KUTLAY, NÜKET, TUNCALI, TİMUR, BEYAZIT, ŞERİFE BÜŞRA, SEMERCİ GÜNDÜZ, CAVİDAN NUR, SIDAR DUMAN, YEŞİM, Ergun, Sezen Guntekin, Ercal, Derya, ALEMDAR, ADEM, ALIYEVA, LAMIYA, ÖZEMRİ SAĞ, ŞEBNEM, Atasever, Umut, AYDIN, ZAFER, Thahir, Adam, TATAR, Abdulgani, ILGIN RUHİ, HATİCE, TERZİ, YUNUS KASIM, BİŞGİN, ATIL, Dincer, Selin Akad, ÖZDEMİR, ÖZTÜRK, ÜLGENALP, AYFER, PERÇİN, FERDA EMRİYE, YILDIRIM, MALİK EJDER, Ulu, Memnune Sena, Solak, Mustafa, Elmas, Muhsin, ÖZDEMİR ERDOĞAN, MÜJGAN, Zararsiz, Gozde Erturk, DEMİR, HÜSEYİN, ÇALIŞ, MUSTAFA, BAŞKOL, MEVLÜT, Aymelek, Huri Sema, ALTINTAŞ, ZUHAL, Eraslan, Serpil, KURT, EMİN EMRE, Erdem, Levent, FAHRİOGLU, UMUT, GÜLEÇ CEYLAN, GÜLAY, Sahin, Izem Olcay, CEYLAN, AHMET CEVDET, TUĞ BOZDOĞAN, SEVCAN, BOĞA, İBRAHİM, Yildiz, Saliha Handan, KARABULUT, HALİL GÜRHAN, YILMAZ, MUSTAFA, TEKEŞ, SELAHADDİN, SILAN, FATMA, KOCABEY, MEHMET, KOÇ, ALTUĞ, ÇANKAYA, TUFAN, BAĞIŞ, HAYDAR, BORA, ELÇİN, GİRAY BOZKAYA, ÖZLEM, ÖZDEMİR, Sevda Yeşim, ÖNAL, MÜGE GÜLCİHAN, ŞENEL, ABDURRAHMAN SONER, POYRAZOĞLU, MUAMMER HAKAN, PAÇ KISAARSLAN, AYŞENUR, GÜRSOY, ŞEBNEM, YÜCE, HÜSEYİN, DUMAN, NİLGÜN, BOZKURT, GÖKAY, Yararbas, Kanay, YILDIRIM, MAHMUT SELMAN, ARMAN, AHMET, MIHÇI, ERCAN, Dicle Üniversitesi, Tıp Fakültesi, Dahili Tıp Bilimleri Bölümü, Tıbbi Genetik Ana Bilim Dalı, Tekeş, Selahaddin, Üzel, Veysiye Hülya, Em, Serda, and DÜNDAR M., FAHRİOGLU U., Yildiz S. H., Bakir-Gungor B., TEMEL Ş. G., AKIN H., ARTAN S., Cora T., ŞAHİN F. İ., DURSUN A., et al.
- Subjects
GENETİK VE KALITIM ,Genotype ,Turkey ,PROTEIN ASC ,MEFV ,Life Sciences (LIFE) ,Molecular Biology and Genetics ,KAPPA-B ,Genotype-phenotype correlations ,Sağlık Bilimleri ,Familial Mediterranean fever ,National Genetics Consortium ,AUTOINFLAMMATION ,ACTIVATION ,Tıbbi Genetik ,Yaşam Bilimleri ,Health Sciences ,Genetics ,Humans ,PYRIN ,GENETICS & HEREDITY ,Molecular Biology ,Moleküler Biyoloji ve Genetik ,Genetics (clinical) ,ASSOCIATIONS ,Internal Medicine Sciences ,MUTATIONS ,Temel Bilimler ,Life Sciences ,General Medicine ,Dahili Tıp Bilimleri ,Tıp ,PREVALENCE ,MOLECULAR BIOLOGY & GENETICS ,Genetics, Population ,Phenotype ,Yaşam Bilimleri (LIFE) ,AMYLOIDOSIS ,Mutation ,Medicine ,Natural Sciences ,Medical Genetics - Abstract
© 2021, The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.Familial Mediterranean fever (FMF) is a monogenic autoinflammatory disorder with recurrent fever, abdominal pain, serositis, articular manifestations, erysipelas-like erythema, and renal complications as its main features. Caused by the mutations in the MEditerranean FeVer (MEFV) gene, it mainly affects people of Mediterranean descent with a higher incidence in the Turkish, Jewish, Arabic, and Armenian populations. As our understanding of FMF improves, it becomes clearer that we are facing with a more complex picture of FMF with respect to its pathogenesis, penetrance, variant type (gain-of-function vs. loss-of-function), and inheritance. In this study, MEFV gene analysis results and clinical findings of 27,504 patients from 35 universities and institutions in Turkey and Northern Cyprus are combined in an effort to provide a better insight into the genotype-phenotype correlation and how a specific variant contributes to certain clinical findings in FMF patients. Our results may help better understand this complex disease and how the genotype may sometimes contribute to phenotype. Unlike many studies in the literature, our study investigated a broader symptomatic spectrum and the relationship between the genotype and phenotype data. In this sense, we aimed to guide all clinicians and academicians who work in this field to better establish a comprehensive data set for the patients. One of the biggest messages of our study is that lack of uniformity in some clinical and demographic data of participants may become an obstacle in approaching FMF patients and understanding this complex disease.
- Published
- 2022
15. Artemisia alleviates AGE-induced liver complications via MAPK and RAGE signaling pathways modulation: a combinatorial study
- Author
-
Hichem Moulahoum, Faezeh Ghorbanizamani, Zineb Khiari, Mohamed Toumi, Yasmina Benazzoug, Kerem Tok, Suna Timur, and Figen Zihnioglu
- Subjects
Glycation End Products, Advanced ,Clinical Biochemistry ,Receptor for Advanced Glycation End Products ,Apoptosis ,Nitric Oxide ,Antioxidants ,Tandem Mass Spectrometry ,Inhibitory-Activity ,Diabetes Mellitus ,Advanced glycation end-products ,Molecular Biology ,Kappa-B ,Flavonoids ,Artemisia herba-alba ,Signaling pathway ,Caspase 3 ,Interleukin-6 ,Plant Extracts ,Tumor Necrosis Factor-alpha ,Diabetes ,Glycation End-Products ,Polyphenols ,Cell Biology ,General Medicine ,Growing Wild ,Glycotoxins ,Essential Oil ,Oxidative Stress ,Artemisia ,Liver ,In-Vitro ,Antioxidant ,Inflammation Mediators ,Proto-Oncogene Proteins c-akt ,Age-related diseases ,Network pharmacology ,Signal Transduction - Abstract
Artemisia herba-alba (AHA) is a traditionally used plant to treat various diseases, including diabetes and metabolic dysfunctions. Plant extracts are generally explored empirically without a deeper assessment of their mechanism of action. Here, we describe a combinatorial study of biochemical, molecular, and bioinformatic (metabolite-protein pharmacology network) analyses to elucidate the mechanism of action of AHA and shed light on its multilevel effects in the treatment of diabetes-related advanced glycation end-products (AGE)-induced liver damages. The extract's polyphenols and flavonoids content were measured and then identified via LC-Q-TOF-MS/MS. Active compounds were used to generate a metabolite-target interaction network via Swiss Target Prediction and other databases. The extract was tested for its antiglycation and aggregation properties. Next, THLE-2 liver cells were challenged with AGEs, and the mechanistic markers were measured [TNF-alpha, IL-6, nitric oxide, total antioxidant capacity, lipid peroxidation (LPO), and caspase 3]. Metabolite and network screening showed the involvement of AHA in diabetes, glycation, liver diseases, aging, and apoptosis. Experimental confirmation showed that AHA inhibited protein modification and AGE formation. Additionally, AHA reduced inflammatory mediators (IL-6, TNF alpha), oxidative stress markers (NO, LPO), and apoptosis (Caspase 3). On the other hand, cellular total antioxidant capacity was restored to normal levels. The combinatorial study showed that AHA regulates AGE-induced liver damages through MAPK-AKT and AGE-RAGE signaling pathways. This report highlights the combination of experimental and network pharmacology for the exact elucidation of AHA mechanism of action as a multitarget option in the therapy of diabetes and AGEs-related diseases., Scientific and Technological Research Council of Turkey-TUBITAK [120Z200]; Projets de Recherche-Formation Universitaire (PRFU) [D01N01UN160420180008], This project was funded by The Scientific and Technological Research Council of Turkey-TUBITAK (120Z200). The current project was partially funded by the Projets de Recherche-Formation Universitaire (PRFU) (D01N01UN160420180008). EGE MATAL (EGE University) is acknowledged for the LC-Q-TOF-MS/MS analyses and cell culture experiments.
- Published
- 2021
16. TBX2, a Novel Regulator of Labour
- Author
-
G. J. M. Veenboer, M. A. M. Kampman, Febilla Fernando, Karst Y. Heida, Louise J M Lagendijk, Gijs B. Afink, Joris A. M. van der Post, Aldo Jongejan, Carrie Ris-Stalpers, Martijn A. Oudijk, Graduate School, Amsterdam Reproduction & Development (AR&D), Obstetrics and Gynaecology, Epidemiology and Data Science, APH - Methodology, Reproductive Biology Laboratory, General Paediatrics, and APH - Personalized Medicine
- Subjects
Chemokine ,Medicine (General) ,BIRTH ,Regulator ,PARTURITION ,KAPPA-B ,progesterone withdrawal ,Article ,Andrology ,Obstetric Labor, Premature ,R5-920 ,Downregulation and upregulation ,INFLAMMATION ,Pregnancy ,Humans ,Medicine ,Transcription factor ,reproductive and urinary physiology ,GENE-EXPRESSION ,Labor, Obstetric ,biology ,Cesarean Section ,Interleukin-6 ,business.industry ,Microarray analysis techniques ,proinflammatory cytokines and chemokines ,myometrium ,TRANSCRIPTION FACTOR TBX2 ,Myometrium ,TBX2 ,PROGESTERONE RECEPTOR-A ,General Medicine ,premature delivery ,HUMAN MYOMETRIAL CELLS ,PLATELET-ACTIVATING-FACTOR ,uterine smooth muscle cells ,CXCL5 ,TERT-HM cells ,biology.protein ,Female ,PRETERM LABOR ,delivery ,T-Box Domain Proteins ,business ,CCL21 - Abstract
Background and Objectives: Therapeutic interventions targeting molecular factors involved in the transition from uterine quiescence to overt labour are not substantially reducing the rate of spontaneous preterm labour. The identification of novel rational therapeutic targets are essential to prevent the most common cause of neonatal mortality. Based on our previous work showing that Tbx2 (T-Box transcription factor 2) is a putative upstream regulator preceding progesterone withdrawal in mouse myometrium, we now investigate the role of TBX2 in human myometrium. Materials and Methods: RNA microarray analysis of (A) preterm human myometrium samples and (B) myometrial cells overexpressing TBX2 in vitro, combined with subsequent analysis of the two publicly available datasets of (C) Chan et al. and (D) Sharp et al. The effect of TBX2 overexpression on cytokines/chemokines secreted to the myometrium cell culture medium were determined by Luminex assay. Results: Analysis shows that overexpression of TBX2 in myometrial cells results in downregulation of TNFα- and interferon signalling. This downregulation is consistent with the decreased expression of cytokines and chemokines of which a subset has been previously associated with the inflammatory pathways relevant for human labour. In contrast, CXCL5 (C-X-C motif chemokine ligand 5), CCL21 and IL-6 (Interleukin 6), previously reported in relation to parturition, do not seem to be under TBX2 control. The combined bioinformatical analysis of the four mRNA datasets identifies a subset of upstream regulators common to both preterm and term labour under control of TBX2. Surprisingly, TBX2 mRNA levels are increased in preterm contractile myometrium. Conclusions: We identified a subset of upstream regulators common to both preterm and term labour that are activated in labour and repressed by TBX2. The increased TBX2 mRNA expression in myometrium collected during a preterm caesarean section while in spontaneous preterm labour compared to tissue harvested during iatrogenic preterm delivery does not fit the bioinformatical model. We can only explain this by speculating that the in vivo activity of TBX2 in human myometrium depends not only on the TBX2 expression levels but also on levels of the accessory proteins necessary for TBX2 activity.
- Published
- 2021
17. BCOR modulates transcriptional activity of a subset of glucocorticoid receptor target genes involved in cell growth and mobility
- Author
-
Jorma J. Palvimo, Einari A. Niskanen, Marjo Malinen, Markku Varjosalo, Joanna K. Lempiäinen, A.B.M. Kaiser Manjur, Institute of Biotechnology, Biosciences, and Molecular Systems Biology
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Biochemistry ,Dexamethasone ,0302 clinical medicine ,Endocrinology ,PROGRAMS ,Cell Movement ,Transcriptional regulation ,Protein Interaction Maps ,Nuclear receptor co-repressor 1 ,Nuclear receptor corepressor 1 (NCOR1) ,Chemistry ,Chromatin binding ,PROLIFERATION ,INHIBITOR ,Transcription regulation ,Coregulator ,Cell biology ,Enhancer Elements, Genetic ,030220 oncology & carcinogenesis ,Molecular Medicine ,Chromatin Immunoprecipitation ,Repressor ,KAPPA-B ,MECHANISMS ,03 medical and health sciences ,Receptors, Glucocorticoid ,INFLAMMATION ,Proto-Oncogene Proteins ,Humans ,Nuclear Receptor Co-Repressor 1 ,Enhancer ,Molecular Biology ,Transcription factor ,Cell Proliferation ,Binding Sites ,COMPLEX ,MUTATIONS ,Glucocorticoid receptor (GR) ,Cell Biology ,BCL6 corepressor (BCOR) ,Repressor Proteins ,HEK293 Cells ,030104 developmental biology ,Gene Expression Regulation ,Nuclear receptor ,MEDULLOBLASTOMA ,1182 Biochemistry, cell and molecular biology ,Corepressor - Abstract
Glucocorticoid (GC) receptor (GR) is a key transcription factor (TF) that regulates vital metabolic and antiinflammatory processes. We have identified BCL6 corepressor (BCOR) as a dexamethasone-stimulated interaction partner of GR. BCOR is a component of non-canonical polycomb repressor complex 1.1 (ncPCR1.1) and linked to different developmental disorders and cancers, but the role of BCOR in GC signaling is poorly characterized. Here, using ChIP-seq we show that, GC induces genome-wide redistribution of BCOR chromatin binding towards GR-occupied enhancers in HEK293 cells. As assessed by RNA-seq, depletion of BCOR altered the expression of hundreds of GC-regulated genes, especially the ones linked to TNF signaling, GR signaling and cell migration pathways. Biotinylation-based proximity mapping revealed that GR and BCOR share several interacting partners, including nuclear receptor corepressor NCOR1. ChIP-seq showed that the NCOR1 co-occurs with both BCOR and GR on a subset of enhancers upon GC treatment. Simultaneous depletion of BCOR and NCOR1 influenced GR target gene expression in a combinatorial and gene-specific manner. Finally, we show using live cell imaging that the depletion of BCOR together with NCOR1 markedly enhances cell migration. Collectively, our data suggest BCOR as an important gene and pathway selective coregulator of GR transcriptional activity.
- Published
- 2021
18. 68Ga-DOTA-E[c(RGDfK)]2 PET Imaging of SHARPIN-Regulated Integrin Activity in Mice
- Author
-
Riikka Siitonen, Anne Roivainen, Meeri Käkelä, Heidi Liljenbäck, Johanna Ivaska, Tiina Saanijoki, Elina Mattila, Anu Autio, Sirpa Jalkanen, Ingrid Dijkgraaf, Emilia Peuhu, Olli Metsälä, RS: CARIM School for Cardiovascular Diseases, Biochemie, RS: Carim - B01 Blood proteins & engineering, and RS: CARIM - R1 - Thrombosis and haemostasis
- Subjects
0301 basic medicine ,EXPRESSION ,alpha(v)beta(3) integrin ,Population ,Patient characteristics ,SHARPIN ,KAPPA-B ,VASCULAR ADHESION PROTEIN-1 ,ANGIOGENESIS ,Continuous variable ,ACTIVATION ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,melanoma ,DOTA ,Medicine ,Radiology, Nuclear Medicine and imaging ,ALPHA-V-BETA-3 ,education ,education.field_of_study ,RGD ,CHRONIC INFLAMMATION ,business.industry ,Blood flow ,Pet imaging ,COMPONENT ,030104 developmental biology ,Heart size ,PET ,chemistry ,Oncology ,Cardiac PET ,030220 oncology & carcinogenesis ,Nuclear medicine ,business ,SKIN - Abstract
1380 Objectives: The purpose of this study was to investigate how gender, BMI, patient cross-sectional area and heart size affect the influence of time-of-flight (TOF) and point spread function (PSF) modeling in image reconstruction on regional absolute myocardial blood flow (MBF) measured with 82Rb PET in an obese population. Methods: Dynamic rest/stress 82Rb cardiac PET studies were performed for 30 obese subjects on a TOF PET scanner. Subject characteristics were (mean ± SD for continuous variables): gender 9 male, 21 female; weight 138.9 ± 32.8 kg; BMI 47.8 ± 9.5 kg/m2; heart size 92.4 ± 55.3 ml; cross-sectional area in a transaxial slice containing the heart 1133 ± 233 cm2. Images were reconstructed in four ways: (1) OSEM without TOF or PSF modeling (OSEM), (2) OSEM with TOF (TOF), (3) OSEM with PSF modeling (PSF) and (4) OSEM with TOF and PSF modeling (PSFTOF). All were with 3 iterations, 8 subsets and with a post-reconstruction 8mm 3D Gaussian filter. A one tissue compartmental model was used to compute MBF. Average rest and stress flow and myocardial flow reserve were computed in five regions: anterior, septal, inferior, lateral and apical. Results from TOF, PSF and PSFTOF were compared with those from OSEM, with subjects grouped by gender, and by tertiles (10 subjects each) for BMI (I: 32.5-40.5, II: 41.1-52.3, III: 53.3-75.6 (kg/m2)), cross-sectional area (I: 532 - 1086, II: 1089 - 1157, III: 1187 - 1662 (cm2)) and heart size (I: 19 -56, II: 66 - 93, III: 97 - 238 (ml)). The MBF or myocardial flow reserve (MFR) in a given region was compared with the corresponding OSEM value and the change was expressed as a percent. Results: The general trends for all subjects with TOF were similar MBF increases in the apical (+22.0%) and septal walls (+19.4%), and a slightly lower increase in the anterior wall (+11.3 %). Increases were small in the inferior (+4.6%) and lateral (+4.9%) walls. There was little effect of PSF modeling in any region. MBF trends with PSFTOF were similar to those for TOF alone. Rest and stress increases with TOF tended to be matched, so that MFR was little changed. Trends were examined for the groupings by patient characteristics. (1) Gender. MBF showed a greater increase with TOF for females (mean +20.9%) compared to males (mean +14.9%) in the apical, septal and anterior walls. Results with PSFTOF were similar. (2) BMI. The MBF increase with TOF in the apical, anterior and septal walls was greatest for tertile II (mean +26.9%) compared with tertile I (mean +13.6%) and tertile III (mean +16.7%). (3) Cross-sectional area. The MBF increase with TOF in the apical, anterior and septal walls was similar for tertiles II (mean +22.1%) and III (mean +22.6%), and was greater than that for tertile I (mean +12.6%). (4) Heart size. The MBF increase with TOF in the apical, anterior and septal walls was greater for smaller heart sizes: tertile I (mean +21.2%), tertile II (+20.7%) and tertile III (+15.3%). These results are for our patient population and particular hardware, software and processing implementation. Conclusions: Patient gender, BMI, cross-sectional area and heart size affect the degree to which MBF values change with TOF. The largest changes are observed for females, patients with larger BMI, larger cross-sectional area and smaller hearts. The greatest changes are found in the apical, septal and lateral walls. The studied patient characteristics have little effect on flow reserve. It is important to consider these patient-dependent effects when evaluating absolute MBF in a clinical environment, and to recognize that equipment or protocol changes may affect MBF results.
- Published
- 2019
19. Therapeutic glucocorticoids prevent bone loss but drive muscle wasting when administered in chronic polyarthritis
- Subjects
VERTEBRAL DEFORMITIES ,RISK ,Polyarthritis ,FRACTURES ,MINERAL DENSITY ,KAPPA-B ,OSTEOPOROSIS ,Glucocorticoids ,Muscle wasting ,RECOMMENDATIONS ,TUMOR-NECROSIS-FACTOR ,RHEUMATOID-ARTHRITIS ,PREVALENCE - Abstract
BackgroundPatients with rheumatoid arthritis (RA) experience extra-articular manifestations including osteoporosis and muscle wasting, which closely associate with severity of disease. Whilst therapeutic glucocorticoids (GCs) reduce inflammation in RA, their actions on muscle and bone metabolism in the context of chronic inflammation remain unclear. We utilised the TNF-tg model of chronic polyarthritis to ascertain the impact of therapeutic GCs on bone and muscle homeostasis in the context of systemic inflammation.MethodsTNF-tg and wild-type (WT) animals received either vehicle or the GC corticosterone (100 mu g/ml) in drinking water at onset of arthritis. Arthritis severity and clinical parameters were measured, serum collected for ELISA and muscle and bone biopsies collected for mu CT, histology and mRNA analysis. In vivo findings were examined in primary cultures of osteoblasts, osteoclasts and myotubes.ResultsTNF-tg mice receiving GCs showed protection from inflammatory bone loss, characterised by a reduction in serum markers of bone resorption, osteoclast numbers and osteoclast activity. In contrast, muscle wasting was markedly increased in WT and TNF-tg animals receiving GCs, independently of inflammation. This was characterised by a reduction in muscle weight and fibre size, and an induction in anti-anabolic and catabolic signalling.ConclusionsThis study demonstrates that when given in early onset chronic polyarthritis, oral GCs partially protect against inflammatory bone loss, but induce marked muscle wasting. These results suggest that in patients with inflammatory arthritis receiving GCs, the development of interventions to manage deleterious side effects in muscle should be prioritised.
- Published
- 2019
20. Dimethyl fumarate treatment in multiple sclerosis
- Subjects
PLACEBO-CONTROLLED PHASE-3 ,B cells ,Adaptive immune system ,Innate immune system ,T cells ,NF-kappa B ,NICOTINIC-ACID ,AUTOIMMUNE ,ACID ESTERS ,KAPPA-B ,Dimethyl fumarate ,Nrf2 ,Multiple sclerosis ,NATURAL-KILLER-CELLS ,MOLECULAR MIMICRY ,B-CELLS ,ORAL BG-12 ,REGULATORY T-CELLS - Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system (CNS) in which demyelination and neurodegeneration occurs. The immune system of MS patients is characterized by a dysregulation in the balance between pro- and anti-inflammatory immune cells, whereby both the innate and adaptive immune system are involved. Dimethyl fumarate (DMF) was licensed in 2013 as an oral first-line therapy for relapsing-remitting (RR)MS patients. It has a strong efficacy with neuroprotective and immunomodulatory effects and a favourable benefit-risk profile. However, the effects of DMF on the immune system of MS patients were not clear before entering the market. During the last years, numerous in vitro and ex vivo studies have clarified the working mechanism of DMF in MS. Here, we discuss the pharmacokinetics of DMF and its effect on molecular immune-related pathways, which is further linked to the clinical and immunological effects of DMF treatment. The efficacy and safety of DMF treatment for RRMS is discussed as reported from clinical trials. Further, the immunological effects of DMF treatment in RRMS patients are addressed in more detail, including the distribution and function of immune cells. Taken together, evidence from recent studies points to a multifactorial working mechanism of DMF treatment in MS which leads to a restored immune balance favouring a more tolerogenic or anti-inflammatory immune profile.
- Published
- 2018
21. Dimethyl fumarate treatment in multiple sclerosis
- Author
-
Gwendoline Montes Diaz, Raymond Hupperts, Judith Fraussen, and Veerle Somers
- Subjects
0301 basic medicine ,Adaptive immune system ,Immunology ,T cells ,NICOTINIC-ACID ,AUTOIMMUNE ,KAPPA-B ,medicine.disease_cause ,Neuroprotection ,Dimethyl fumarate ,Nrf2 ,Multiple sclerosis ,03 medical and health sciences ,chemistry.chemical_compound ,NATURAL-KILLER-CELLS ,MOLECULAR MIMICRY ,0302 clinical medicine ,Immune system ,ORAL BG-12 ,medicine ,Animals ,Humans ,Immunology and Allergy ,REGULATORY T-CELLS ,Autoimmune disease ,PLACEBO-CONTROLLED PHASE-3 ,B cells ,Innate immune system ,business.industry ,NF-kappa B ,ACID ESTERS ,Prognosis ,medicine.disease ,Acquired immune system ,Molecular mimicry ,030104 developmental biology ,chemistry ,B-CELLS ,business ,Immunosuppressive Agents ,030217 neurology & neurosurgery - Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system (CNS) in which demyelination and neurodegeneration occurs. The immune system of MS patients is characterized by a dysregulation in the balance between pro- and anti-inflammatory immune cells, whereby both the innate and adaptive immune system are involved. Dimethyl fumarate (DMF) was licensed in 2013 as an oral first-line therapy for relapsing-remitting (RR)MS patients. It has a strong efficacy with neuroprotective and immunomodulatory effects and a favourable benefit-risk profile. However, the effects of DMF on the immune system of MS patients were not clear before entering the market. During the last years, numerous in vitro and ex vivo studies have clarified the working mechanism of DMF in MS. Here, we discuss the pharmacokinetics of DMF and its effect on molecular immune-related pathways, which is further linked to the clinical and immunological effects of DMF treatment. The efficacy and safety of DMF treatment for RRMS is discussed as reported from clinical trials. Further, the immunological effects of DMF treatment in RRMS patients are addressed in more detail, including the distribution and function of immune cells. Taken together, evidence from recent studies points to a multifactorial working mechanism of DMF treatment in MS which leads to a restored immune balance favouring a more tolerogenic or anti-inflammatory immune profile.
- Published
- 2018
22. Towards a Better Understanding of Cohesin Mutations in AML
- Author
-
Sergi Cuartero, Andrew J. Innes, Matthias Merkenschlager, Wellcome Trust, Medical Research Council (MRC), and NIHR
- Subjects
0301 basic medicine ,Cancer Research ,Cohesin complex ,Mini Review ,cohesin ,KAPPA-B ,Biology ,lcsh:RC254-282 ,BONE-MARROW-CELLS ,03 medical and health sciences ,0302 clinical medicine ,AML ,SISTER-CHROMATID COHESION ,medicine ,HEMATOPOIETIC STEM-CELLS ,1112 Oncology and Carcinogenesis ,GENE-EXPRESSION ,Cohesin ,Inflammation ,Science & Technology ,Leukemia ,leukemia ,Myeloid leukemia ,interferon ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,MYELOID-LEUKEMIA CELLS ,Phenotype ,hematopoiesis ,Cell biology ,Chromatin ,Hematopoiesis ,REMODELING COMPLEX ,Establishment of sister chromatid cohesion ,SELF-RENEWAL ,Haematopoiesis ,030104 developmental biology ,EPIGENETIC REGULATORS ,PROGENITOR CELLS ,Oncology ,inflammation ,030220 oncology & carcinogenesis ,Interferon ,Life Sciences & Biomedicine - Abstract
Classical driver mutations in acute myeloid leukemia (AML) typically affect regulators of cell proliferation, differentiation, and survival. The selective advantage of increased proliferation, improved survival, and reduced differentiation on leukemia progression is immediately obvious. Recent large-scale sequencing efforts have uncovered numerous novel AML-associated mutations. Interestingly, a substantial fraction of the most frequently mutated genes encode general regulators of transcription and chromatin state. Understanding the selective advantage conferred by these mutations remains a major challenge. A striking example are mutations in genes of the cohesin complex, a major regulator of three-dimensional genome organization. Several landmark studies have shown that cohesin mutations perturb the balance between self-renewal and differentiation of hematopoietic stem and progenitor cells (HSPC). Emerging data now begin to uncover the molecular mechanisms that underpin this phenotype. Among these mechanisms is a role for cohesin in the control of inflammatory responses in HSPCs and myeloid cells. Inflammatory signals limit HSPC self-renewal and drive HSPC differentiation. Consistent with this, cohesin mutations promote resistance to inflammatory signals, and may provide a selective advantage for AML progression. In this review, we discuss recent progress in understanding cohesin mutations in AML, and speculate whether vulnerabilities associated with these mutations could be exploited therapeutically.
- Published
- 2021
23. Can endolysosomal deacidification and inhibition of autophagy prevent severe COVID-19?
- Author
-
Morris, Gerwyn, Athan, Eugene, Walder, Ken, Bortolasci, Chiara C., O'Neil, Adrienne, Marx, Wolf, Berk, Michael, Carvalho, André F., Maes, Michael, Puri, Basant K., Morris, Gerwyn, Athan, Eugene, Walder, Ken, Bortolasci, Chiara C., O'Neil, Adrienne, Marx, Wolf, Berk, Michael, Carvalho, André F., Maes, Michael, and Puri, Basant K.
- Published
- 2020
24. Early vascular ageing in chronic kidney disease: impact of inflammation, vitamin K, senescence and genomic damage
- Author
-
Dai, Lu, Dai, Lu, Schurgers, Leon J., Shiels, Paul G., Stenvinkel, Peter, Dai, Lu, Dai, Lu, Schurgers, Leon J., Shiels, Paul G., and Stenvinkel, Peter
- Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by cardiovascular disease, persistent uraemic inflammation, osteoporosis muscle wasting and frailty. The accelerated early vascular ageing (EVA) process mediated by medial vascular calcification (VC) is a hallmark of senescence as well as a strong predictor of cardiovascular morbidity and mortality in the CKD population. Current clinical therapeutic strategies and novel treatments for VC have not yet been proven to prevent or reverse VC progression in patients with CKD. Knowledge of the fundamental mechanism underlying EVA is urgently needed to identify and develop novel and efficient therapeutic targets for VC and EVA. An accumulating body of evidence indicates that deoxyribonucleic acid (DNA) damage-induced cellular senescence and 'inflammaging' may largely contribute to such pathological conditions characterized by accelerated EVA. Growing evidence shows that nuclear factor erythroid 2-related factor 2 (NRF2) signalling and vitamin K play a crucial role in counteracting oxidative stress, DNA damage, senescence and inflammaging, whereby NRF2 activation and vitamin K supplementation may provide a novel treatment target for EVA. In this review we discuss the link between senescence and EVA in the context of CKD, with a focus on the role of NRF2 and vitamin K in DNA damage signalling, senescence and inflammaging.
- Published
- 2020
25. Long Non-Coding RNAs Involved in Progression of Non-Alcoholic Fatty Liver Disease to Steatohepatitis
- Author
-
Bart van de Sluis, Folkert Kuipers, Ronit Shiri-Sverdlov, Cisca Wijmenga, Jingyuan Fu, Glenn Marsman, Biljana Atanasovska, Sander S. Rensen, Sebo Withoff, Molecular Neuroscience and Ageing Research (MOLAR), Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Center for Liver, Digestive and Metabolic Diseases (CLDM), Restoring Organ Function by Means of Regenerative Medicine (REGENERATE), Surgery, RS: NUTRIM - R2 - Liver and digestive health, and Moleculaire Genetica
- Subjects
0301 basic medicine ,Cirrhosis ,PATHOGENESIS ,Non-alcoholic Fatty Liver Disease/genetics ,LIPOTOXICITY ,Fatty Acids, Nonesterified ,Chronic liver disease ,Transcriptome ,long non-coding RNAs ,Fatty Acids, Nonesterified/pharmacology ,Long Noncoding/genetics ,ENDOPLASMIC-RETICULUM STRESS ,Liver/drug effects ,RNA-Seq ,Biology (General) ,Fatty Acids ,NF-kappa B/genetics ,Fatty liver ,NF-kappa B ,Hep G2 Cells ,General Medicine ,3. Good health ,Liver ,Tumor Necrosis Factor-alpha/pharmacology ,Disease Progression ,RNA, Long Noncoding ,Tumor necrosis factor alpha ,Inflammation Mediators ,Hepatocytes/drug effects ,Liver cancer ,functional genomics ,HEPATIC INFLAMMATION ,QH301-705.5 ,PATHOPHYSIOLOGY ,KAPPA-B ,Biology ,Article ,MECHANISMS ,03 medical and health sciences ,medicine ,Humans ,Gene silencing ,RNA, Long Noncoding/genetics ,RECEPTOR ,030102 biochemistry & molecular biology ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,non-alcoholic fatty liver disease ,medicine.disease ,digestive system diseases ,030104 developmental biology ,Renal disorders Radboud Institute for Molecular Life Sciences [Radboudumc 11] ,Case-Control Studies ,Nonesterified/pharmacology ,Hepatocytes ,Cancer research ,RNA ,Steatohepatitis - Abstract
Contains fulltext : 238435.pdf (Publisher’s version ) (Open Access) Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease and is characterized by different stages varying from benign fat accumulation to non-alcoholic steatohepatitis (NASH) that may progress to cirrhosis and liver cancer. In recent years, a regulatory role of long non-coding RNAs (lncRNAs) in NAFLD has emerged. Therefore, we aimed to characterize the still poorly understood lncRNA contribution to disease progression. Transcriptome analysis in 60 human liver samples with various degrees of NAFLD/NASH was combined with a functional genomics experiment in an in vitro model where we exposed HepG2 cells to free fatty acids (FFA) to induce steatosis, then stimulated them with tumor necrosis factor alpha (TNFα) to mimic inflammation. Bioinformatics analyses provided a functional prediction of novel lncRNAs. We further functionally characterized the involvement of one novel lncRNA in the nuclear-factor-kappa B (NF-κB) signaling pathway by its silencing in Hepatoma G2 (HepG2) cells. We identified 730 protein-coding genes and 18 lncRNAs that responded to FFA/TNFα and associated with human NASH phenotypes with consistent effect direction, with most being linked to inflammation. One novel intergenic lncRNA, designated lncTNF, was 20-fold up-regulated upon TNFα stimulation in HepG2 cells and positively correlated with lobular inflammation in human liver samples. Silencing lncTNF in HepG2 cells reduced NF-κB activity and suppressed expression of the NF-κB target genes A20 and NFKBIA. The lncTNF we identified in the NF-κB signaling pathway may represent a novel target for controlling liver inflammation.
- Published
- 2021
26. Betacyanins, major components in Opuntia red-purple fruits, protect against acetaminophen-induced acute liver failure
- Author
-
Klaas Nico Faber, Pieter G. Tepper, Ma Consolación Martínez-Saldaña, Han Moshage, Wim J. Quax, Herson Antonio González-Ponce, Manon Buist-Homan, Chemical and Pharmaceutical Biology, Biopharmaceuticals, Discovery, Design and Delivery (BDDD), Nanotechnology and Biophysics in Medicine (NANOBIOMED), Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Center for Liver, Digestive and Metabolic Diseases (CLDM), and Restoring Organ Function by Means of Regenerative Medicine (REGENERATE)
- Subjects
Antioxidant ,NAPQI ,030309 nutrition & dietetics ,HEPATOTOXICITY ,medicine.medical_treatment ,Nutraceutical compounds ,BETALAIN ,KAPPA-B ,Opuntia fruit extracts ,METABOLISM ,medicine.disease_cause ,HEPATOCYTES ,03 medical and health sciences ,chemistry.chemical_compound ,0404 agricultural biotechnology ,Nutraceutical ,medicine ,Betacyanins ,Betanin ,Acetaminophen ,Liver injury ,0303 health sciences ,Traditional medicine ,Plant Extracts ,INDUCTION ,Opuntia ,ANTIOXIDANT PROPERTIES ,04 agricultural and veterinary sciences ,Liver Failure, Acute ,medicine.disease ,GADD45-BETA ,040401 food science ,chemistry ,Oxidative stress ,Fruit ,JNK ,Acute liver failure ,Food Science ,medicine.drug - Abstract
Acetaminophen (APAP) misuse or overdose is the most important cause of drug-induced acute liver failure. Overdoses of acetaminophen induce oxidative stress and liver injury by the electrophilic metabolite N-acetyl-pbenzoquinone imine (NAPQI). Plant-based medicine has been used for centuries against diseases or intoxications due to their biological activities. The aim of this study was to evaluate the therapeutic value of Opuntia robusta and Opuntia streptacantha fruit extracts against acetaminophen-induced liver damage and to identify the major biocomponents on them. Opuntia fruit extracts were obtained by peeling and squeezing each specie, followed by lyophilization. HPLC was used to characterize the extracts. The effect of the extracts against acetaminophen induced acute liver injury was evaluated both in vivo and in vitro using biochemical, molecular and histological determinations. The results showed that betacyanins are the main components in the analyzed Opuntia fruit extracts, with betanin as the highest concentration. Therapeutic treatments with Opuntia extracts reduced biochemical, molecular and histological markers of liver (in vivo) and hepatocyte (in vitro) injury. Opuntia extracts reduced the APAP-increased expression of the stress-related gene Gadd45b. Furthermore, Opuntia extracts exerted diverse effects on the antioxidant related genes Sod2, Gclc and Hmox1, independent of their ROSscavenging ability. Therefore, betacyanins as betanin from Opuntia robusta and Opuntia streptacantha fruits are promising nutraceutical compounds against oxidative liver damage.
- Published
- 2020
27. Adipocyte calcium sensing receptor is not involved in visceral adipose tissue inflammation or atherosclerosis development in hyperlipidemic Apoe
- Author
-
Yvonne Jansen, Christian Weber, Sai Sahana Sundararaman, Florian Kahles, Michael Lehrke, Selin Gencer, Andrea Bonnin Marquez, Emiel P. C. van der Vorst, Linsey J. F. Peters, Yi Yan, Sumra Nazir, Yvonne Döring, Joachim Jankowski, Erik A.L. Biessen, Pathologie, RS: Carim - B07 The vulnerable plaque: makers and markers, Biochemie, and RS: Carim - B01 Blood proteins & engineering
- Subjects
0301 basic medicine ,Mice, Knockout, ApoE ,Adipose tissue ,030204 cardiovascular system & hematology ,Systemic inflammation ,DISEASE ,ACTIVATION ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adipocyte ,Adipocytes ,610 Medicine & health ,Receptor ,TUMOR-NECROSIS-FACTOR ,Multidisciplinary ,PROLIFERATION ,Plaque, Atherosclerotic ,Mechanisms of disease ,OBESITY ,Medicine ,Tumor necrosis factor alpha ,medicine.symptom ,Calcium-sensing receptor ,EXTRACELLULAR CA2+-SENSING RECEPTOR ,EXPRESSION ,medicine.medical_specialty ,Science ,PARATHYROID-HORMONE ,Inflammation ,Hyperlipidemias ,KAPPA-B ,Intra-Abdominal Fat ,Article ,03 medical and health sciences ,Internal medicine ,medicine ,INTERLEUKIN-6 PRODUCTION ,Animals ,Humans ,business.industry ,Cardiovascular biology ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,chemistry ,business ,Receptors, Calcium-Sensing ,Homeostasis - Abstract
Scientific reports 11, 10409 (2021). doi:10.1038/s41598-021-89893-y, Published by Macmillan Publishers Limited, part of Springer Nature, [London]
- Published
- 2020
28. Progressive Skeletal Muscle Atrophy in Muscular Dystrophies: A Role for Toll-like Receptor-Signaling in Disease Pathogenesis
- Author
-
Boel De Paepe
- Subjects
Muscle tissue ,DOWN-REGULATION ,DUCHENNE ,Inflammation ,Review ,KAPPA-B ,Protein degradation ,UP-REGULATION ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,Downregulation and upregulation ,SERUM BIOMARKERS ,Medicine and Health Sciences ,Medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,DEFICIENT SKELETAL ,Receptor ,Muscle, Skeletal ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,GENE-EXPRESSION ,Toll-like receptor ,business.industry ,Organic Chemistry ,INFLAMMATORY RESPONSE ,UBIQUITIN-PROTEASOME ,General Medicine ,MOUSE MODEL ,Muscle atrophy ,Cell biology ,Computer Science Applications ,Toll-like receptors ,TLR2 ,Muscular Atrophy ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Disease Progression ,muscular dystrophies ,medicine.symptom ,business ,myositis ,Signal Transduction - Abstract
Muscle atrophy is an active process controlled by specific transcriptional programs, in which muscle mass is lost by increased protein degradation and/or decreased protein synthesis. This review explores the involvement of Toll-like receptors (TLRs) in the muscle atrophy as it is observed in muscular dystrophies, disorders characterized by successive bouts of muscle fiber degeneration and regeneration in an attempt to repair contraction-induced damage. TLRs are defense receptors that detect infection and recognize self-molecules released from damaged cells. In muscular dystrophies, these receptors become over-active, and are firmly involved in the sustained chronic inflammation exhibited by the muscle tissue, via their induction of pro-inflammatory cytokine expression. Taming the exaggerated activation of TLR2/4 and TLR7/8/9, and their downstream effectors in particular, comes forward as a therapeutic strategy with potential to slow down disease progression.
- Published
- 2020
29. Pathways involved in viral oncogenesis: New perspectives from virus-host protein interactomics
- Author
-
Kazim Yalcin Arga, Medi Kori, Kori, Medi, and Arga, Kazim Yalcin
- Subjects
0301 basic medicine ,CERVICAL-CANCER ,Signaling pathways ,Carcinogenesis ,viruses ,Viral oncogenesis ,SIGNAL-TRANSDUCTION ,Context (language use) ,KAPOSIS-SARCOMA ,KAPPA-B ,Biology ,medicine.disease_cause ,Oncoviruses ,Virus ,03 medical and health sciences ,Viral Proteins ,0302 clinical medicine ,Neoplasms ,Protein Interaction Mapping ,medicine ,CELL-CYCLE ,Humans ,Protein Interaction Maps ,EPSTEIN-BARR-VIRUS ,Molecular Biology ,TUMOR-NECROSIS-FACTOR ,HEPATITIS-C-VIRUS ,Retinoblastoma ,SARCOMA-ASSOCIATED HERPESVIRUS ,Cell cycle ,medicine.disease ,Androgen receptor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Virus-host protein interactions ,Host-Pathogen Interactions ,Cancer research ,Molecular Medicine ,Signal transduction ,Oncogenic Viruses ,ALPHA-MEDIATED APOPTOSIS ,Oncovirus ,Signal Transduction - Abstract
Oncogenic viruses are among the apparent causes of cancer-associated mortality. It was estimated that 12% to 15% of human malignancies are linked to oncoviruses. Although modernist strategies and traditional genetic studies have defined host-pathogen interactions of the oncoviruses, their host functions which are critical for the establishment of infection still remain mysterious. However, over the last few years, it has become clear that infections hijack and modify cellular pathways for their benefit. In this context, we constructed the virus-host protein interaction networks of seven oncoviruses (EBV, HBV, HCV, HTLV-1, HHV8, HPV16, and HPV18), and revealed cellular pathways hijacking as a result of oncogenic virus infection. Several signaling pathways/processes such as TGF-beta signaling, cell cycle, retinoblastoma tumor suppressor protein, and androgen receptor signaling were mutually targeted by viruses to induce oncogenesis. Besides, cellular pathways specific to a certain virus were detected. By this study, we believe that we improve the understanding of the molecular pathogenesis of viral oncogenesis and provide information in setting new targets for treatment, prognosis, and diagnosis.
- Published
- 2020
30. Dietary Advanced Glycation Endproducts Decrease Glucocorticoid Sensitivity In Vitro
- Author
-
Antoon Opperhuizen, Timme van der Lugt, Aalt Bast, Misha F. Vrolijk, Antje R. Weseler, Farmacologie en Toxicologie, RS: NUTRIM - R3 - Respiratory & Age-related Health, RS: FSE UCV Adaptive responses in relation to health effect and safety of nutrition, and FSE Campus Venlo
- Subjects
Glycation End Products, Advanced ,0301 basic medicine ,THP-1 Cells ,Drug Resistance ,Pharmacology ,medicine.disease_cause ,ACTIVATION ,0302 clinical medicine ,Glucocorticoid receptor ,oxidative stress ,Phosphorylation ,chemistry.chemical_classification ,reactive oxygen species ,Nutrition and Dietetics ,Chemistry ,theophylline ,030220 oncology & carcinogenesis ,medicine.symptom ,lcsh:Nutrition. Foods and food supply ,hormones, hormone substitutes, and hormone antagonists ,Intracellular ,END-PRODUCTS ,ACETYLATION ,kappa-b ,INHIBITION ,lcsh:TX341-641 ,Inflammation ,inflammatory bowel diseases ,Article ,03 medical and health sciences ,Receptors, Glucocorticoid ,Glucocorticoid Sensitivity ,glucocorticoid resistance ,medicine ,Humans ,Interleukin 8 ,Glucocorticoids ,Reactive oxygen species ,receptor phosphorylation ,Macrophages ,Interleukin-8 ,gene-expression ,Maillard Reaction ,030104 developmental biology ,inflammation ,dietary advanced glycation endproducts ,CELLS ,Oxidative stress ,Food Science - Abstract
Glucocorticoids are very effective anti-inflammatory drugs and widely used for inflammatory bowel disease (IBD) patients. However, approximately 20% of IBD patients do not respond to glucocorticoids and the reason for this is largely unknown. Dietary advanced glycation endproducts (AGEs) are formed via the Maillard reaction during the thermal processing of food products and can induce a pro-inflammatory reaction in human cells. To investigate whether this pro-inflammatory response could be mitigated by glucocorticoids, human macrophage-like cells were exposed to both LPS and AGEs to induce interleukin-8 (IL8) secretion. This pro-inflammatory response was then modulated by adding pharmacological compounds interfering in different steps of the anti-inflammatory mechanism of glucocorticoids: rapamycin, quercetin, and theophylline. Additionally, intracellular reactive oxygen species (ROS) were measured and the glucocorticoid receptor phosphorylation state was assessed. The results show that AGEs induced glucocorticoid resistance, which could be mitigated by quercetin and rapamycin. No change in the phosphorylation state of the glucocorticoid receptor was observed. Additionally, intracellular ROS formation was induced by AGEs, which was mitigated by quercetin. This suggests that AGE-induced ROS is an underlying mechanism to AGE-induced glucocorticoid resistance. This study shows for the first time the phenomenon of dietary AGE-induced glucocorticoid resistance due to the formation of ROS. Our findings indicate that food products with a high inflammatory potential can induce glucocorticoid resistance, these results may be of great importance to IBD patients suffering from glucocorticoid resistance.
- Published
- 2020
31. Dexamethasone Inhibits Spheroid Formation of Thyroid Cancer Cells Exposed to Simulated Microgravity
- Author
-
Melnik, Daniela, Sahana, Jayashree, Corydon, Thomas J, Kopp, Sascha, Nassef, Mohamed Zakaria, Wehland, Markus, Infanger, Manfred, Grimm, Daniela, and Krüger, Marcus
- Subjects
ANCHORAGE-INDEPENDENT GROWTH ,MULTICELLULAR TUMOR ,glucocorticoids ,proliferation ,FACTOR-BETA ,3D growth ,epithelial-mesenchymal transition ,KAPPA-B ,epithelial–mesenchymal transition ,Article ,PROSTATE-CANCER ,LUNG-CANCER ,lcsh:Biology (General) ,anoikis ,E-CADHERIN ,EXTRACELLULAR-MATRIX ,nuclear factor kappa-light-chain-enhancer of activated b-cells (nf-κb) ,nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-kappa B) ,3d growth ,lcsh:QH301-705.5 ,GENE-EXPRESSION - Abstract
Detachment and the formation of spheroids under microgravity conditions can be observed with various types of intrinsically adherent human cells. In particular, for cancer cells this process mimics metastasis and may provide insights into cancer biology and progression that can be used to identify new drug/target combinations for future therapies. By using the synthetic glucocorticoid dexamethasone (DEX), we were able to suppress spheroid formation in a culture of follicular thyroid cancer (FTC)-133 cells that were exposed to altered gravity conditions on a random positioning machine. DEX inhibited the growth of three-dimensional cell aggregates in a dose-dependent manner. In the first approach, we analyzed the expression of several factors that are known to be involved in key processes of cancer progression such as autocrine signaling, proliferation, epithelial−mesenchymal transition, and anoikis. Wnt/β-catenin signaling and expression patterns of important genes in cancer cell growth and survival, which were further suggested to play a role in three-dimensional aggregation, such as NFKB2, VEGFA, CTGF, CAV1, BCL2(L1), or SNAI1, were clearly affected by DEX. Our data suggest the presence of a more complex regulation network of tumor spheroid formation involving additional signal pathways or individual key players that are also influenced by DEX.
- Published
- 2020
32. MANF ablation causes prolonged activation of the UPR without neurodegeneration in the mouse midbrain dopamine system
- Author
-
Mart Saarma, Piotr Chmielarz, Petteri Piepponen, Maria Lindahl, Mikko Airavaara, Emmi Pakarinen, Vootele Voikar, Tatiana Danilova, Institute of Biotechnology, Biosciences, Neuroscience Center, Regenerative pharmacology group, Drug Research Program, Timo Petteri Piepponen / Principal Investigator, University Management, Division of Pharmacology and Pharmacotherapy, Divisions of Faculty of Pharmacy, Helsinki One Health (HOH), Staff Services, Mart Saarma / Principal Investigator, and Organotypic Vasculature Lab
- Subjects
Male ,Endoribonuclease activity ,Substantia nigra ,KAPPA-B ,NEUROTROPHIC FACTOR MANF ,Biology ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,Mice ,0302 clinical medicine ,ENDOPLASMIC-RETICULUM STRESS ,PARKINSONS-DISEASE ,Dopamine ,Neurotrophic factors ,Mesencephalon ,Endoribonucleases ,medicine ,Animals ,IMMUNOREACTIVITY ,Nerve Growth Factors ,030304 developmental biology ,MANF ,0303 health sciences ,ATF6 ,UNFOLDED PROTEIN RESPONSE ,General Neuroscience ,Neurodegeneration ,3112 Neurosciences ,General Medicine ,ER STRESS ,medicine.disease ,GENE ,NERVOUS-SYSTEM ,Cell biology ,ALZHEIMERS-DISEASE ,nervous system ,knock-out mice ,Knockout mouse ,Unfolded protein response ,Disorders of the Nervous System ,Female ,CNS ,dopamine ,030217 neurology & neurosurgery ,Research Article: New Research ,medicine.drug - Abstract
Visual Abstract, Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) localized protein that regulates ER homeostasis and unfolded protein response (UPR). The biology of endogenous MANF in the mammalian brain is unknown and therefore we studied the brain phenotype of MANF-deficient female and male mice at different ages focusing on the midbrain dopamine system and cortical neurons. We show that a lack of MANF from the brain led to the chronic activation of UPR by upregulation of the endoribonuclease activity of the inositol-requiring enzyme 1α (IRE1α) pathway. Furthermore, in the aged MANF-deficient mouse brain in addition the protein kinase-like ER kinase (PERK) and activating transcription factor 6 (ATF6) branches of the UPR pathways were activated. Neuronal loss in neurodegenerative diseases has been associated with chronic ER stress. In our mouse model, increased UPR activation did not lead to neuronal cell loss in the substantia nigra (SN), decrease of striatal dopamine or behavioral changes of MANF-deficient mice. However, cortical neurons lacking MANF were more vulnerable to chemical induction of additional ER stress in vitro. We conclude that embryonic neuronal deletion of MANF does not cause the loss of midbrain dopamine neurons in mice. However, endogenous MANF is needed for maintenance of neuronal ER homeostasis both in vivo and in vitro.
- Published
- 2020
33. The secretome derived from mesenchymal stromal cells cultured in a xeno-free medium promotes human cartilage recovery in vitro
- Author
-
Maria Elisabetta Federica Palamà, Georgina Margaret Shaw, Simonetta Carluccio, Daniele Reverberi, Laura Sercia, Luana Persano, Dario Pisignano, Katia Cortese, Francis Peter Barry, Josephine Mary Murphy, Chiara Gentili, Horizon 2020, Science Foundation Ireland, and Seventh Framework Programme
- Subjects
EXPRESSION ,0301 basic medicine ,Histology ,Stromal cell ,MATRIX METALLOPROTEINASES ,lcsh:Biotechnology ,Biomedical Engineering ,Mesenchymal stromal cells ,Arthritis ,Bioengineering ,KAPPA-B ,02 engineering and technology ,Osteoarthritis ,Matrix metalloproteinase ,Andrology ,Extracellular matrix ,03 medical and health sciences ,lcsh:TP248.13-248.65 ,EXTRACELLULAR-MATRIX ,medicine ,extracellular vesicles ,inflammation ,mesenchymal stromal cells ,osteoarthritis ,secretome ,xeno-free medium ,Original Research ,Secretome ,Inflammation ,Xeno-free medium ,Chemistry ,Mesenchymal stem cell ,Bioengineering and Biotechnology ,ARTICULAR-CARTILAGE ,Extracellular vesicles ,021001 nanoscience & nanotechnology ,medicine.disease ,In vitro ,3. Good health ,DIFFERENTIATION ,030104 developmental biology ,ARTHRITIS ,Stem cell ,BONE ,0210 nano-technology ,STEM-CELLS ,Biotechnology - Abstract
Osteoarthritis (OA) is a disabling joint disorder causing articular cartilage degeneration. Currently, the treatments are mainly aimed to pain and symptoms relief, rather than disease amelioration. Human bone marrow stromal cells (hBMSCs) have emerged as a promising paracrine mechanism-based tool for OA treatment. Here, we investigate the therapeutic potential of conditioned media (CM) and extracellular vesicles (EVs) isolated from hBMSC and grown in a xeno-free culture system (XFS) compared to the conventional fetal bovine serum-culture system (FBS) in an in vitro model of OA. First, we observed that XFS promoted growth and viability of hBMSCs compared to FBS-containing medium while preserving their typical phenotype. The biological effects of the CM derived from hBMSC cultivated in XFS- and FBS-based medium were tested on IL-1 alpha treated human chondrocytes, to mimic the OA enviroment. Treatment with CM derived from XFS-cultured hBMSC inhibited IL-1 alpha-induced expression of IL-6, IL-8, and COX-2 by hACs compared to FBS-based condition. Furthermore, we observed that hBMSCs grown in XFS produced a higher amount of EVs compared to FBS-culture. The hBMSC-EVs not only inhibit the adverse effects of IL-1 alpha-induced inflammation, but play a significant in vitro chondroprotective effect. In conclusion, the XFS medium was found to be suitable for isolation and expansion of hBMSCs with increased safety profile and intended for ready-to-use clinical therapies. This work was received funding from the European Union’s Horizon 2020 Research and Innovation Program under grant agreement AUTOSTEM number 667932, the Science Foundation Ireland grant number 13/TIDA/B2668, and the FP7 European Union Health Collaborative Project PurStem, grant number 223298. peer-reviewed
- Published
- 2020
34. Identification of regulatory variants associated with genetic susceptibility to meningococcal disease
- Author
-
Borghini, Lisa, Png, Eileen, Binder, Alexander, Wright, Victoria J, Pinnock, Ellie, de Groot, Ronald, Hazelzet, Jan, Emonts, Marieke, Van der Flier, Michiel, Schlapbach, Luregn J, Anderson, Suzanne, Secka, Fatou, Salas, Antonio, Fink, Colin, Carrol, Enitan D, Pollard, Andrew J, Coin, Lachlan J, Kuijpers, Taco W, Martinon-Torres, Federico, Zenz, Werner, Levin, Michael, Hibberd, Martin L, Davila, Sonia, Gormley, Stuart, Hamilton, Shea, Herberg, Jethro, Hourmat, Bernardo, Hoggart, Clive, Kaforou, Myrsini, Sancho-Shimizu, Vanessa, Abdulla, Amina, Agapow, Paul, Bartlett, Maeve, Bellos, Evangelos, Eleftherohorinou, Hariklia, Galassini, Rachel, Inwald, David, Mashbat, Meg, Menikou, Stefanie, Mustafa, Sobia, Nadel, Simon, Rahman, Rahmeen, Thakker, Clare, Bokhandi, S, Power, Sue, Barham, Heather, Pathan, N, Ridout, Jenna, White, Deborah, Thurston, Sarah, Faust, S, Patel, S, McCorkell, Jenni, Davies, P, Cratev, Lindsey, Navarra, Helen, Carter, Stephanie, Ramaiah, R, Patel, Rekha, Tuffrey, Catherine, Gribbin, Andrew, McCready, Sharon, Peters, Mark, Hardy, Katie, Standing, Fran, O'Neill, Lauren, Abelake, Eugenia, Deep, Akash, Nsirim, Eniola, Willis, Louise, Young, Zoe, Royad, C, White, Sonia, Fortune, PM, Hudnott, Phil, Alvez Gonzalez, Fernando, Barral-Arca, Ruth, Cebey-Lopez, Miriam, Jose Curras-Tuala, Maria, Garcia, Natalia, Garcia Vicente, Luisa, Gomez-Carballa, Alberto, Gomez Rial, Jose, Grela Beiroa, Andrea, Justicia Grande, Antonio, Leborans Iglesias, Pilar, Martinez Santos, Alba Elena, Martinon-Torres, Nazareth, Martinon Sanchez, Jose Maria, Mosquera Perez, Belen, Obando Pacheco, Pablo, Pardo-Seco, Jacobo, Pischedda, Sara, Rivero Calle, Irene, Rodriguez-Tenreiro, Carmen, Redondo-Collazo, Lorenzo, Seren Fernandez, Sonia, Porto Silva, Maria del Sol, Vega, Ana, Beatriz Reyes, Susana, Leon Leon, Maria Cruz, Navarro Mingorance, Alvaro, Gabaldo Barrios, Xavier, Onate Vergara, Eider, Concha Torre, Andres, Vivanco, Ana, Fernandez, Reyes, Gimenez Sanchez, Francisco, Sanchez Forte, Miguel, Rojo, Pablo, Ruiz Contreras, J, Palacios, Alba, Navarro, Marisa, Alvarez Alvarez, Cristina, Jose Lozano, Maria, Carreras, Eduardo, Brio Sanagustin, Sonia, Neth, Olaf, Martinez Padilla, Ma del Carmen, Prieto Tato, Luis Manuel, Guillen, Sara, Fernandez Silveira, Laura, Moreno, David, van Furth, AM Tutu, van der Flier, M, Boeddha, NP, Driessen, GJA, Pajkrt, D, Sanders, EAM, van de Beek, D, van der Ende, A, Philipsen, HLA, Adeel, AOA, Breukels, MA, Brinkman, DMC, de Korte, CCMM, de Vries, E, de Waal, WJ, Dekkers, R, Dings-Lammertink, A, Doedens, RA, Donker, AE, Dousma, M, Faber, TE, Gerrits, GPJM, Gerver, JAM, Heidema, J, Homan-van der Veen, J, Jacobs, MAM, Jansen, NJG, Kawczynski, P, Klucovska, K, Kneyber, MCJ, Koopman-Keemink, Y, Langenhorst, VJ, Leusink, J, Loza, BF, Merth, IT, Miedema, CJ, Neeleman, C, Noordzij, JG, Obihara, CC, van Overbeek-van Gils, ALT, Poortman, GH, Potgieter, ST, Potjewijd, J, Rosias, PPR, Sprong, T, ten Tussher, GW, Thio, BJ, Tramper-Stranders, GA, van Deuren, M, van der Meer, H, van Kuppevelt, AJM, van Wermeskerken, AM, Verwijs, WA, Wolfs, TFW, Agyeman, Philipp, Aebi, Christoph, Berger, Christoph, Giannoni, Eric, Stocker, Martin, Posfay-Barbe, Klara M, Heininger, Ulrich, Bernhard-Stirnemann, Sara, Niederer-Loher, Anita, Kahlert, Christian, Hasters, Paul, Relly, Christa, Baer, Walter, Paulus, Stephane, Frederick, Hannah, Jennings, Rebecca, Johnston, Joanne, Kenwright, Rhian, Agbeko, Rachel, Bojang, Kalifa, Sarr, Isatou, Kebbeh, Ngane, Sey, Gibbi, Saidykhan, Momodou, Cole, Fatoumatta, Thomas, Gilleh, Antonio, Martin, Walcher, Wolfgang, Geishofer, Gotho, Klobassa, Daniela, Martin, Mueller, Pfurtscheller, Klaus, Reiter, Karl, Roedl, Siegfried, Zobel, Gerfried, Zoehrer, Bettina, Toepke, Baerbel, Fucik, Peter, Gabriel, Markwart, Penzien, Johann M, Diab, Gedeon, Miething, Robert, Deeg, KH, Hammer, Jurg, Varnholt, Verena, Schmidt, Andreas, Bindl, Lutz, Sillaber, Ursula, Huemer, Christian, Meier, Primrose, Simic-Schleicher, G, Markart, Markus, Pfau, Eberhard, Broede, Hans, Ausserer, Bernd, Kalhoff, Hermann, Arpe, Volker, Schweitzer-Krantz, Susanne, Kasper, Johannes-Martin, Loranth, Kathrin, Bittrich, Hans J, Simma, Burkhard, Klinge, Jens, Fedlmaier, Michael, Weigand, Nicola, Herting, Egbert, Grube, Regina, Fusch, Christoph, Gruber, Alois, Schimmel, Ulf, Knaufer-Schiefer, Suzanne, Laessig, Wolfgang, Hennenberger, Axel, von der Wense, Axel, Tillmann, Roland, Schwarick, Juergen, Sitzmann, Friedrich C, Streif, Werner, Mueller, Herbert, Kurnik, Peter, Groneck, Peter, Weiss, Ute, Groeblacher-Roth, Helene, Bensch, Juergen, Moser, Reinhard, Schwarz, Rudolf, Lenz, Kurt, Hofmann, Thomas, Goepel, Wolfgang, Schulz, Dietrich, Berger, Thomas, Hauser, Erwin, Foerster, Kai Martin, Peters, Jochen, Nicolai, T Homas, Kumlien, Bjoern, Beckmann, Regina, Seitz, Christiane, Hueseman, D, Schuermann, Roland, Ta, Van Hop, Weikmann, Eckart, Evert, W, Hautz, Juergen, Seidenberg, Juergen, Wocko, Lucia, Luigs, Petra, Reiter, Hans-Ludwig, Quietzach, J, Koenig, Michael, Herrmann, Johanna, Mitter, Horst, Seidler, Ekkehard, Maak, Bernhard, Sperl, Wolfgang, Zwiauer, Karl, Meissl, Manfred, Koch, Reinhard, Cremer, Manfred, Breuer, HA, Goerke, W, Nossal, Robert, Pernice, Walter, Brangenberg, Ralf, Salzer, Hans R, Koch, Hartmut, Schaller, Gerhard, Paky, Franz, Strasser, Friedrich, Eitelberger, Franz, Sontheimer, D, Lischka, Andreas, Kronberger, Martina, Dilch, Alfred, Scheibenpflug, Christian, Bruckner, Robert, Mahler, Klaus, Runge, Klaus, Kunze, Wolfgang, Schermann, Peter, Consortium, EUCLIDS, European Commission, Meningitis Research Foundation, National Institute for Health Research (UK), Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne Hospitals NHS Foundation Trust, Instituto de Salud Carlos III, Xunta de Galicia, Wyeth Farma, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela, Novartis, Swiss National Science Foundation, Swiss Society of Intensive Care Medicine, Gottfried und Julia Bangerter-Rhyner-Stiftung, Vinetum and Borer Foundation, Foundation for the Health of Children and Adolescents, Austrian National Bank, University of Zurich, Borghini, Lisa, Universidade de Santiago de Compostela. Departamento de Ciencias Forenses, Anatomía Patolóxica, Xinecoloxía e Obstetricia, e Pediatría, Png, Eileen [0000-0001-5586-6395], Wright, Victoria J [0000-0001-7826-1516], Schlapbach, Luregn J [0000-0003-2281-2598], Salas, Antonio [0000-0002-2336-702X], Martinon-Torres, Federico [0000-0002-9023-581X], Apollo - University of Cambridge Repository, ARD - Amsterdam Reproduction and Development, AII - Infectious diseases, Paediatric Infectious Diseases / Rheumatology / Immunology, Neurology, ANS - Neuroinfection & -inflammation, Medical Microbiology and Infection Prevention, Pediatrics, Public Health, Posfay Barbe, Klara, Critical care, Anesthesiology, Peri-operative and Emergency medicine (CAPE), APH - Aging & Later Life, Amsterdam Reproduction & Development (AR&D), and Pediatric surgery
- Subjects
0301 basic medicine ,Candidate gene ,Hypopharyngeal Neoplasms / microbiology ,Meningococcal Infections / microbiology ,Vascular damage Radboud Institute for Health Sciences [Radboudumc 16] ,MathematicsofComputing_GENERAL ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,lcsh:Medicine ,Neisseria meningitidis ,Regulatory Sequences, Nucleic Acid ,Cohort Studies ,0302 clinical medicine ,ComputingMethodologies_SYMBOLICANDALGEBRAICMANIPULATION ,Hypopharyngeal Neoplasms / pathology ,BINDING ,Tumor Cells, Cultured ,lcsh:Science ,Genetics ,Multidisciplinary ,ddc:618 ,Cultured ,TheoryofComputation_GENERAL ,High-Throughput Nucleotide Sequencing ,Genomics ,Phenotype ,3. Good health ,ddc ,Tumor Cells ,Multidisciplinary Sciences ,GENOME ,Science & Technology - Other Topics ,Neisseria meningitidis / isolation & purification ,Single-nucleotide polymorphism ,610 Medicine & health ,KAPPA-B ,Biology ,Hypopharyngeal Neoplasms / genetics ,Polymorphism, Single Nucleotide ,Article ,Meningococcal Infections / epidemiology ,03 medical and health sciences ,SEQUENCE VARIATION ,Genetic predisposition ,Journal Article ,Humans ,Meningitis ,Meningococcal Infections / genetics ,Genetic Predisposition to Disease ,Nucleic Acid ,Polymorphism ,General ,Gene ,Genotyping ,Neisseria meningitidis / genetics ,Genetic Association Studies ,Genetic association ,Genetic association study ,1000 Multidisciplinary ,Science & Technology ,Hypopharyngeal Neoplasms ,genetic variants ,meningococcal disease ,lcsh:R ,Single Nucleotide ,EUCLIDS consortium ,Gene regulation ,Meningococcal Infections ,030104 developmental biology ,10036 Medical Clinic ,Case-Control Studies ,lcsh:Q ,Software_PROGRAMMINGLANGUAGES ,Regulatory Sequences ,030217 neurology & neurosurgery - Abstract
EUCLIDS consortium., Non-coding genetic variants play an important role in driving susceptibility to complex diseases but their characterization remains challenging. Here, we employed a novel approach to interrogate the genetic risk of such polymorphisms in a more systematic way by targeting specific regulatory regions relevant for the phenotype studied. We applied this method to meningococcal disease susceptibility, using the DNA binding pattern of RELA – a NF-kB subunit, master regulator of the response to infection – under bacterial stimuli in nasopharyngeal epithelial cells. We designed a custom panel to cover these RELA binding sites and used it for targeted sequencing in cases and controls. Variant calling and association analysis were performed followed by validation of candidate polymorphisms by genotyping in three independent cohorts. We identified two new polymorphisms, rs4823231 and rs11913168, showing signs of association with meningococcal disease susceptibility. In addition, using our genomic data as well as publicly available resources, we found evidences for these SNPs to have potential regulatory effects on ATXN10 and LIF genes respectively. The variants and related candidate genes are relevant for infectious diseases and may have important contribution for meningococcal disease pathology. Finally, we described a novel genetic association approach that could be applied to other phenotypes., This work has been partially funded by the European Seventh Framework Programme for Research and Technological Development (FP7) under EUCLIDS Grant Agreement No. 279185. The UK meningococcal cohort was established with support from Meningitis Research Foundation through grants to Imperial College London. The Research from Newcastle partners was supported by the National Institute for Health Research Newcastle Biomedical Research Centre based at Newcastle Hospitals NHS Foundation Trust and Newcastle University. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health. The ESIGEM Research group activities were supported by grants from Instituto de Salud Carlos III (Proyecto de Investigación en Salud, Acción Estratégica en Salud: proyecto GePEM PI16/01478) (A.S.); Consellería de Sanidade, Xunta de Galicia (RHI07/2-intensificación actividad investigadora, PS09749 and 10PXIB918184PR), Instituto de Salud Carlos III (Intensificación de la actividad investigadora 2007–2016), Convenio de colaboración de investigación (Wyeth España-Fundación IDICHUS 2007–2011), Convenio de colaboración de investigación (Novartis España-Fundación IDICHUS 2010–2011), Fondo de Investigación Sanitaria (FIS; PI070069/PI1000540) del plan nacional de I + D + I and ‘fondos FEDER’ (F.M.T.). The Swiss Pediatric Sepsis study was funded by grants from the Swiss National Science Foundation (342730_153158/1), the Swiss Society of Intensive Care, the Bangerter Foundation, the Vinetum and Borer Foundation, and the Foundation for the Health of Children and Adolescents. The Western Europe Meningococcal Study was supported by grants no 8842, 10112 and 12710 of the Oesterreichische Nationalbank (Austria), grants A3-16.K-8/2008-11 and A3-16.K-8/2006–9 of the Department for Science and Research of the Styrian federal government (Austria) and the non for profit association ‘In Vita’, Graz (Austria).
- Published
- 2019
35. Altered protein turnover signaling and myogenesis during impaired recovery of inflammation-induced muscle atrophy in emphysematous mice
- Author
-
Stefan J. van Hoof, Annemie M. W. J. Schols, Magda M Drożdż, Ramon C. J. Langen, Anita Kneppers, Frank Verhaegen, Roger P H A Rosenbrand, Chiel C. de Theije, Judith J.M. Ceelen, Marco C. J. M. Kelders, RS: NUTRIM - R3 - Respiratory & Age-related Health, Promovendi NTM, Pulmonologie, RS: CAPHRI - R2 - Creating Value-Based Health Care, Health Services Research, Ondersteunend personeel NTM, RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, and Radiotherapie
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,SYSTEMIC INFLAMMATION ,lcsh:Medicine ,Inflammation ,KAPPA-B ,MyoD ,Muscle Development ,OBSTRUCTIVE PULMONARY-DISEASE ,Article ,EXERCISE CAPACITY ,03 medical and health sciences ,Myoblast fusion ,Mice ,0302 clinical medicine ,Internal medicine ,SATELLITE CELLS ,medicine ,Myocyte ,Animals ,Muscle, Skeletal ,lcsh:Science ,Multidisciplinary ,Myogenesis ,business.industry ,lcsh:R ,Protein turnover ,Skeletal muscle ,Recovery of Function ,MOUSE MODEL ,Muscle atrophy ,Mice, Inbred C57BL ,Muscular Atrophy ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,030228 respiratory system ,Pulmonary Emphysema ,MYOBLAST FUSION ,EXACERBATION ,SKELETAL-MUSCLE ,lcsh:Q ,medicine.symptom ,business ,MYOD ,Signal Transduction - Abstract
Exacerbations in Chronic obstructive pulmonary disease (COPD) are often accompanied by pulmonary and systemic inflammation, and are associated with an increased susceptibility to weight loss and muscle wasting. As the emphysematous phenotype in COPD appears prone to skeletal muscle wasting, the aims of this study were to evaluate in emphysematous compared to control mice following repetitive exacerbations (1) changes in muscle mass and strength and, (2) whether muscle mass recovery and its underlying processes are impaired. Emphysema was induced by intra-tracheal (IT) elastase instillations, followed by three weekly IT-LPS instillations to mimic repetitive exacerbations. Loss of muscle mass and strength were measured, and related to analyses of muscle protein turnover and myogenesis signaling in tissue collected during and following recovery. Emphysematous mice showed impaired muscle mass recovery in response to pulmonary inflammation-induced muscle atrophy. Proteolysis and protein synthesis signaling remained significantly higher in emphysematous mice during recovery from LPS. Myogenic signaling in skeletal muscle was altered, and fusion capacity of cultured muscle cells treated with plasma derived from LPS-treated emphysematous mice was significantly decreased. In conclusion, repetitive cycles of pulmonary inflammation elicit sustained muscle wasting in emphysematous mice due to impaired muscle mass recovery, which is accompanied by aberrant myogenesis.
- Published
- 2018
36. Structure–function analyses of the bacterial zinc metalloprotease effector protein GtgA uncover key residues required for deactivating NF-κB
- Author
-
Elliott Jennings, Teresa L. M. Thurston, Katrin Rittinger, Diego Esposito, and Wellcome Trust
- Subjects
MECHANISM ,0301 basic medicine ,bacterial pathogenesis ,metalloprotease ,substrate specificity ,medicine.disease_cause ,Biochemistry ,virulence factor ,NF-κB ,Type three secretion system ,type III secretion system (T3SS) ,chemistry.chemical_compound ,SUBSTRATE RECOGNITION ,SPECIFICITY ,chemistry.chemical_classification ,Metalloproteinase ,REFINEMENT ,Effector ,RELB ,Escherichia coli Proteins ,bacterial effectors ,NF-kappa B ,Salmonella enterica ,11 Medical And Health Sciences ,Cell biology ,Amino acid ,Zinc ,ESCHERICHIA-COLI ,SECRETION SYSTEM ,03 Chemical Sciences ,Life Sciences & Biomedicine ,NF-B ,Biochemistry & Molecular Biology ,KAPPA-B ,03 medical and health sciences ,NLEC ,Bacterial Proteins ,medicine ,Editors' Picks ,Molecular Biology ,Escherichia coli ,Transcription factor ,Science & Technology ,GtgA ,Cell Biology ,DNA ,06 Biological Sciences ,030104 developmental biology ,chemistry ,Metalloproteases ,HOMODIMER - Abstract
The closely related type III secretion system zinc metalloprotease effector proteins GtgA, GogA, and PipA are translocated into host cells during Salmonella infection. They then cleave nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) transcription factor subunits, dampening activation of the NF-κB signaling pathway and thereby suppressing host immune responses. We demonstrate here that GtgA, GogA, and PipA cleave a subset of NF-κB subunits, including p65, RelB, and cRel but not NF-κB1 and NF-κB2, whereas the functionally similar type III secretion system effector NleC of enteropathogenic and enterohemorrhagic Escherichia coli cleaved all five NF-κB subunits. Mutational analysis of NF-κB subunits revealed that a single nonconserved residue in NF-κB1 and NF-κB2 that corresponds to the P1′ residue Arg-41 in p65 prevents cleavage of these subunits by GtgA, GogA, and PipA, explaining the observed substrate specificity of these enzymes. Crystal structures of GtgA in its apo-form and in complex with the p65 N-terminal domain explained the importance of the P1′ residue. Furthermore, the pattern of interactions suggested that GtgA recognizes NF-κB subunits by mimicking the shape and negative charge of the DNA phosphate backbone. Moreover, structure-based mutational analysis of GtgA uncovered amino acids that are required for the interaction of GtgA with p65, as well as those that are required for full activity of GtgA in suppressing NF-κB activation. This study therefore provides detailed and critical insight into the mechanism of substrate recognition by this family of proteins important for bacterial virulence.
- Published
- 2018
37. Co-Activation of Glucocorticoid Receptor and Peroxisome Proliferator–Activated Receptor-γ in Murine Skin Prevents Worsening of Atopic March
- Author
-
Hamida Hammad, Viacheslav Mylka, Marnik Vuylsteke, Michael Devos, Bart N. Lambrecht, Lode De Cauwer, Sofie Desmet, Giel Tanghe, Jan Tavernier, Justine Van Moorleghem, Nadia Bougarne, Manon Vanheerswynghels, Karolien De Bosscher, Jonathan Thommis, Astrid Luypaert, and Julie Deckers
- Subjects
0301 basic medicine ,Peroxisome proliferator-activated receptor ,Inflammation ,KAPPA-B ,Dermatology ,DENDRITIC CELLS ,Biochemistry ,Allergic sensitization ,03 medical and health sciences ,Glucocorticoid receptor ,PPAR-ALPHA ,Medicine and Health Sciences ,HOUSE-DUST MITE ,medicine ,Molecular Biology ,chemistry.chemical_classification ,House dust mite ,biology ,business.industry ,Biology and Life Sciences ,ALLERGIC SENSITIZATION ,GM-CSF ,Cell Biology ,Atopic dermatitis ,TRANSACTIVATION ,medicine.disease ,biology.organism_classification ,respiratory tract diseases ,3. Good health ,DERMATITIS ,030104 developmental biology ,chemistry ,Nuclear receptor ,MOUSE BONE-MARROW ,Immunology ,AGGRAVATES EXPERIMENTAL ASTHMA ,medicine.symptom ,business ,Glucocorticoid ,medicine.drug - Abstract
Children with atopic dermatitis show an increased risk to develop asthma later in life, a phenomenon referred to as "atopic march," which emphasizes the need for secondary prevention therapies. This study aimed to investigate whether relief of skin inflammation by glucocorticoids and peroxisome proliferator-activated receptor agonists might influence the subsequent development of asthma in a murine model for the atopic march in which mice were repeatedly exposed to house dust mite via the skin, followed by exposure to house dust mite in lungs. To abrogate atopic dermatitis, mice received topical treatment with glucocorticoid receptor/peroxisome proliferator-activated receptor-gamma agonists. Nuclear receptor ligand effects were assessed on primary keratinocytes and dendritic cells, as central players in skin inflammation. Prior house dust mite-induced skin inflammation aggravates allergic airway inflammation and induces a mixed T helper type 2/T helper type 17 response in the lungs. Cutaneous combined activation of glucocorticoid receptor/peroxisome proliferator-activated receptor-gamma reduced skin inflammation to a higher extent compared to single activation. Additive anti-inflammatory effects were more prominent in dendritic cells, as compared to keratinocytes. Alleviation of allergic skin inflammation by activation of glucocorticoid receptor/peroxisome proliferator-activated receptor-gamma appeared insufficient to avoid the allergic immune response in the lungs, but efficiently reduced asthma severity by counteracting the Th17 response. Glucocorticoid receptor/peroxisome proliferator-activated receptor-gamma co-activation represents a potent remedy against allergic skin inflammation and worsening of atopic march.
- Published
- 2018
38. N-Acetyl-3-aminopyrazoles block the non-canonical NF-kB cascade by selectively inhibiting NIK
- Author
-
Stefano Sainas, Jean-Marie Contreras, Alessandro Barge, Salam Al-Karadaghi, Alex Ducime, Agnese Chiara Pippione, Marco Lucio Lolli, Antonella Federico, Elisa Lupino, Michael Kubbutat, Christophe Morice, Marco Piccinini, and Donatella Boschi
- Subjects
0301 basic medicine ,Drug target ,Pharmaceutical Science ,KAPPA-B ,Pyrazole ,Inhibitory postsynaptic potential ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,KAPPA-B, BREAST-CANCER, MULTIPLE-MYELOMA, aminopyrazoles, NF-kB cascade, NIK inhibitor ,MULTIPLE-MYELOMA ,Drug Discovery ,BREAST-CANCER ,Gene ,Pharmacology ,Kinase ,Organic Chemistry ,NF-kB cascade ,aminopyrazoles ,NIK inhibitor ,3. Good health ,Cell biology ,030104 developmental biology ,Non canonical ,chemistry ,Molecular Medicine - Abstract
NF-κB-inducing kinase (NIK), an oncogenic drug target that is associated with various cancers, is a central signalling component of the non-canonical pathway. A blind screening process, which established that amino pyrazole related scaffolds have an effect on IKKbeta, led to a hit-to-lead optimization process that identified the aminopyrazole 3a as a low μM selective NIK inhibitor. Compound 3a effectively inhibited the NIK-dependent activation of the NF-κB pathway in tumour cells, confirming its selective inhibitory profile.
- Published
- 2018
39. Disease-Associated SNPs in Inflammation-Related lncRNAs
- Author
-
Genética, antropología física y fisiología animal, Genetika,antropologia fisikoa eta animalien fisiologia, Castellanos Rubio, Ainara, Ghosh, Sankar, Genética, antropología física y fisiología animal, Genetika,antropologia fisikoa eta animalien fisiologia, Castellanos Rubio, Ainara, and Ghosh, Sankar
- Abstract
Immune-mediated diseases, such as celiac disease, type 1 diabetes or multiple sclerosis, are a clinically heterogeneous group of diseases that share many key genetic triggers. Although the pathogenic mechanisms responsible for the development of immune mediated disorders is not totally understood, high-throughput genomic studies, such as GWAS and Immunochip, performed in the past few years have provided intriguing hints about underlying mechanisms and pathways that lead to disease. More than a hundred gene variants associated with disease susceptibility have been identified through such studies, but the progress toward understanding the underlying mechanisms has been slow. The majority of the identified risk variants are located in non-coding regions of the genome making it difficult to assign a molecular function to the SNPs. However, recent studies have revealed that many of the non-coding regions bearing disease-associated SNPs generate long non-coding RNAs (lncRNAs). LncRNAs have been implicated in several inflammatory diseases, and many of them have been shown to function as regulators of gene expression. Many of the disease associated SNPs located in lncRNAs modify their secondary structure, or influence expression levels, thereby affecting their regulatory function, hence contributing to the development of disease.
- Published
- 2019
40. The PCSK9-LDL Receptor Axis and Outcomes in Heart Failure BIOSTAT-CHF Subanalysis
- Subjects
EXPRESSION ,DECREASES ,MORTALITY ,CHOLESTEROL ,heart failure ,KAPPA-B ,DEGRADATION ,PCSK9 ,LDLR ,low-density lipoprotein receptor ,proprotein convertase subtilisin/kexin type 9 ,INHIBITORS ,ROSUVASTATIN ,CONVERTASE SUBTILISIN/KEXIN TYPE-9 - Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds low-density lipoprotein receptor (LDLR), preventing its recycling. PCSK9 is a risk predictor and a biotarget in atherosclerosis progression.OBJECTIVES The aim of this study was to determine whether the PCSK9-LDLR axis could predict risk in patients with heart failure (HF).METHODS The BIOSTAT-CHF (Biology Study to Tailored Treatment in Chronic Heart Failure) is a multicenter, multinational, prospective, observational study that included patients with worsening HF signs and/or symptoms. The primary endpoints were all-cause mortality and the composite of mortality or unscheduled hospitalizations for HF. We implemented Cox proportional hazard regression to determine the simultaneously adjusted effect of PCSK9 and LDLR on both outcomes when added to the previously validated BIOSTAT-CHF risk scores.RESULTS This study included 2,174 patients (mean age: 68 +/- 12 years; 53.2% had a history of ischemic heart disease). Median (interquartile range) PCSK9 and LDLR levels were 1.81 U/ml (1.45 to 2.18) and 2.98 U/ml (2.45 to 3.53), respectively. During follow-up, 569 deaths (26.2%) and 896 (41.2%) composite endpoints were ascertained. A multivariable analysis, which included BIOSTAT-CHF risk scores, LDLR, and statin treatment as covariates, revealed a positive linear association between PCSK9 levels and the risk of mortality (hazard ratio [HR]: 1.24; 95% confidence interval [CI]: 1.04 to 1.49; p = 0.020) and the composite endpoint (HR: 1.21; 95% CI: 1.05 to 1.40; p = 0.010). A similar analysis for LDLR revealed a negative association with mortality (HR: 0.86; 95% CI: 0.76 to 0.98; p = 0.025) and the composite endpoint (HR: 0.92; 95% CI: 0.83 to 1.01; p = 0.087). Including PCSK9 and LDLR improved risk score performance.CONCLUSIONS The PCSK9-LDLR axis was associated with outcomes in patients with HF. Future studies must assess whether PCSK9 inhibition will result in better outcomes in HF.
- Published
- 2017
41. Macrophage complexity in human atherosclerosis
- Subjects
polarization ,arteriosclerosis ,RECEPTOR ,PREDICT CARDIOVASCULAR EVENTS ,UNSTABLE ANGINA-PECTORIS ,macrophage ,KAPPA-B ,PLAQUE VULNERABILITY ,ACTIVATION ,MYOCARDIAL-INFARCTION ,CAROTID-ENDARTERECTOMY ,rupture ,monocytes ,innate immunity ,MONOCYTE SUBSETS ,STATIN TREATMENT - Abstract
Purpose of reviewThe pivotal role of macrophages in experimental atherosclerosis is firmly established, but their contribution to human disease is less well defined. In this review we have outlined the current insights on macrophage phenotypes and their presumed precursors, monocytes, in clinical atherosclerosis, and their association with disease progression. Moreover, we will assess major clinical modifiers of macrophage-mediated plaque inflammation and define the outstanding questions for further study.Recent findingsOur survey indicates that macrophage accumulation and status in human plaques are linked with lesion progression and destabilization as well as with symptomatic coronary artery disease. Likewise, levels of their precursors, circulating monocytes were repeatedly seen to associate with atherosclerosis and to predict clinical outcome. Furthermore, the presence and phenotype of both macrophages and monocytes appears to be responsive to the traditional risk factors of atherosclerosis, including hypercholesterolemia, hypertension, and type 2 diabetes, and to treatment thereof, with clear repercussions on disease development,SummaryAlthough plaque macrophages and their precursor cells do represent attractive targets for treating cardiovascular diseases, this therapeutic avenue requires much deeper understanding of the complexity of macrophage biology in human atherosclerosis than available at present.
- Published
- 2017
42. Macrophage complexity in human atherosclerosis
- Author
-
Kristiaan Wouters and Erik A.L. Biessen
- Subjects
0301 basic medicine ,Experimental atherosclerosis ,PREDICT CARDIOVASCULAR EVENTS ,Endocrinology, Diabetes and Metabolism ,macrophage ,KAPPA-B ,030204 cardiovascular system & hematology ,ACTIVATION ,03 medical and health sciences ,0302 clinical medicine ,Human disease ,Risk Factors ,Genetics ,Animals ,Humans ,Medicine ,Macrophage ,innate immunity ,Molecular Biology ,polarization ,Nutrition and Dietetics ,Innate immune system ,Monocyte subsets ,arteriosclerosis ,RECEPTOR ,business.industry ,Macrophages ,Disease progression ,UNSTABLE ANGINA-PECTORIS ,Cell Biology ,Arteriosclerosis ,Statin treatment ,Atherosclerosis ,medicine.disease ,PLAQUE VULNERABILITY ,030104 developmental biology ,MYOCARDIAL-INFARCTION ,Immunology ,Disease Progression ,CAROTID-ENDARTERECTOMY ,rupture ,monocytes ,Cardiology and Cardiovascular Medicine ,business ,MONOCYTE SUBSETS ,STATIN TREATMENT - Abstract
Purpose of review The pivotal role of macrophages in experimental atherosclerosis is firmly established, but their contribution to human disease is less well defined. In this review we have outlined the current insights on macrophage phenotypes and their presumed precursors, monocytes, in clinical atherosclerosis, and their association with disease progression. Moreover, we will assess major clinical modifiers of macrophage-mediated plaque inflammation and define the outstanding questions for further study. Recent findings Our survey indicates that macrophage accumulation and status in human plaques are linked with lesion progression and destabilization as well as with symptomatic coronary artery disease. Likewise, levels of their precursors, circulating monocytes were repeatedly seen to associate with atherosclerosis and to predict clinical outcome. Furthermore, the presence and phenotype of both macrophages and monocytes appears to be responsive to the traditional risk factors of atherosclerosis, including hypercholesterolemia, hypertension, and type 2 diabetes, and to treatment thereof, with clear repercussions on disease development, Summary Although plaque macrophages and their precursor cells do represent attractive targets for treating cardiovascular diseases, this therapeutic avenue requires much deeper understanding of the complexity of macrophage biology in human atherosclerosis than available at present.
- Published
- 2017
43. Involvement of c-Jun N-Terminal Kinase in TNF-alpha-Driven Remodeling
- Author
-
Cheryl van de Wetering, Yvonne M. W. Janssen-Heininger, Emiel F.M. Wouters, Jos van der Velden, Evi M. Mercken, Scott W. Aesif, Rafael de Cabo, Niki L. Reynaert, Irene M. J. Eurlings, Mieke A. Dentener, Promovendi NTM, Pulmonologie, RS: NUTRIM - R3 - Respiratory & Age-related Health, RS: NUTRIM - R3 - Chronic inflammatory disease and wasting, and MUMC+: MA Longziekten (3)
- Subjects
0301 basic medicine ,Pathology ,Clinical Biochemistry ,Vimentin ,Matrix metalloproteinase ,UP-REGULATION ,Mesoderm ,Extracellular matrix ,Mice ,Pulmonary Disease, Chronic Obstructive ,Phosphorylation ,c-Jun N-terminal kinase ,Original Research ,TUMOR-NECROSIS-FACTOR ,c-jun ,respiratory system ,Pulmonary Surfactant-Associated Protein C ,EPITHELIAL-MESENCHYMAL TRANSITION ,Cell Hypoxia ,Extracellular Matrix ,Cell biology ,Phenotype ,medicine.symptom ,Signal Transduction ,TNF-alpha ,Pulmonary and Respiratory Medicine ,EXPRESSION ,medicine.medical_specialty ,Inflammation ,KAPPA-B ,Biology ,OBSTRUCTIVE PULMONARY-DISEASE ,Cell Line ,lung ,03 medical and health sciences ,INFLAMMATION ,medicine ,Animals ,Humans ,ALVEOLAR ,Epithelial–mesenchymal transition ,TGF-BETA-1 ,Molecular Biology ,Tumor Necrosis Factor-alpha ,JNK Mitogen-Activated Protein Kinases ,Epithelial Cells ,Surfactant protein C ,Cell Biology ,respiratory tract diseases ,030104 developmental biology ,CELLS ,biology.protein ,Elastin ,matrix remodeling ,Biomarkers - Abstract
Lung tissue remodeling in chronic obstructive pulmonary disease (COPD) is characterized by airway wall thickening and/or emphysema. Although the bronchial and alveolar compartments are functionally independent entities, we recently showed comparable alterations in matrix composition comprised of decreased elastin content and increased collagen and hyaluronan contents of alveolar and small airway walls. Out of several animal models tested, surfactant protein C (SPC)-TNF-alpha mice showed remodeling in alveolar and airway walls similar to what we observed in patients with COPD. Epithelial cells are able to undergo a phenotypic shift, gaining mesenchymal properties, a process in which c-Jun N-terminal kinase (JNK) signaling is involved. Therefore, we hypothesized that TNF-alpha induces JNK-dependent epithelial plasticity, which contributes to lung matrix remodeling. To this end, the ability of TNF-alpha to induce a phenotypic shift was assessed in A549, BEAS2B, and primary bronchial epithelial cells, and phenotypic markers were studied in SPC-TNF-alpha mice. Phenotypic markers of mesenchymal cells were elevated both in vitro and in vivo, as shown by the expression of vimentin, plasminogen activator inhibitor-1, collagen, and matrix metalloproteinases. Concurrently, the expression of the epithelial markers, E-cadherin and keratin 7 and 18, was attenuated. A pharmacological inhibitor of JNK attenuated this phenotypic shift in vitro, demonstrating involvement of JNK signaling in this process. Interestingly, activation of JNK signaling was also clearly present in lungs of SPC-TNF-alpha mice and patients with COPD. Together, these data show a role for TNF-alpha in the induction of a phenotypic shift in vitro, resulting in increased collagen production and the expression of elastin-degrading matrix metalloproteinases, and provide evidence for involvement of the TNF-alpha-JNK axis in extracellular matrix remodeling.
- Published
- 2017
44. Paraquat disrupts the anti-inflammatory action of cortisol in human macrophages in vitro: therapeutic implications for paraquat intoxications
- Author
-
Antje R. Weseler, Gesiele Veríssimo, Aalt Bast, Farmacologie en Toxicologie, RS: NUTRIM - R3 - Respiratory & Age-related Health, RS: CARIM - R3.05 - Vascular remodeling in cardiovascular disease, and RS: NUTRIM - R3 - Chronic inflammatory disease and wasting
- Subjects
GLUCOCORTICOID RESISTANCE ,0301 basic medicine ,medicine.drug_class ,Health, Toxicology and Mutagenesis ,INDUCED OXIDATIVE STRESS ,HERBICIDE PARAQUAT ,Inflammation ,KAPPA-B ,Pharmacology ,Toxicology ,OBSTRUCTIVE PULMONARY-DISEASE ,Anti-inflammatory ,ACUTE LUNG INJURY ,03 medical and health sciences ,chemistry.chemical_compound ,Paraquat ,NADPH OXIDASE ,medicine ,TRANSCRIPTION FACTOR ,Interleukin 8 ,Viability assay ,INDUCED NEUROTOXICITY ,business.industry ,Glutathione ,Chemistry ,CORTICOSTEROID RESISTANCE ,030104 developmental biology ,chemistry ,Toxicity ,Tumor necrosis factor alpha ,medicine.symptom ,business - Abstract
The herbicide paraquat (1,1'-dimethyl-4,4'-bipyridinium dichloride) has been banned in Europe since 2007 due to its high toxicity in humans. However, it is still widely used in Middle/ South America and in Asia where it is annually associated with a high incidence of unintentional and intentional poisoning. Human macrophage-like cell lines were used to shed more light on the inflammatory response elicited by paraquat. Paraquat (3-1000 mu M) reduced cell viability in a dose-and time-dependent manner. Exposure to 50 or 200 mu M paraquat for 24 h elevated the release of interleukin 8 and gene expression of tumor necrosis factor-alpha. Expression of the 11 beta-hydroxysteroid dehydrogenase 1 gene tended to increase, while cellular glutathione concentrations decreased. The anti-inflammatory effect of cortisol was significantly disrupted. The paraquat-induced cortisol resistance could not be prevented by N-acetyl-L-cysteine. However, a polyphenolic extract of grape seeds consisting of monomeric and oligomeric flavan-3-ols (MOF) reduced paraquat-induced inflammation in the presence of cortisol to baseline. In conclusion, the results suggest that an impaired cortisol response may contribute to paraquat-mediated inflammation. Agents with pleiotropic cellular and subcellular effects on redox regulation and inflammation, such as plant-derived polyphenols, may be an effective add-on to the therapy of paraquat intoxications with glucocorticoids.
- Published
- 2017
45. Colon cancer cell-derived 12(S)-HETE induces the retraction of cancer-associated fibroblast via MLC2, RHO/ROCK and Ca2+ signalling
- Author
-
Julia Eichsteininger, Konstantin Alexander Brendel, Helga Schachner, Mira Stadler, Benedikt Giessrigl, Wolfgang M. Schmidt, Oskar Koperek, Daniel Senfter, Chi Huu Nguyen, Georg Krupitza, Walter Jäger, Serena Stadler, Daniela Milovanovic, Zsuzsanna Bago-Horvath, Nicole Huttary, Helmut Dolznig, Brigitte Marian, Robert M. Mader, Stefan Brenner, Sigurd Krieger, Silvio Holzner, Liselotte Krenn, and Olivier De Wever
- Subjects
0301 basic medicine ,Myosin light-chain kinase ,Stromal cell ,CARCINOMA-CELLS ,KAPPA-B ,Signal transduction ,Biology ,COLORECTAL-CANCER ,Extracellular matrix ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Tumour progression ,0302 clinical medicine ,Stroma ,Medicine and Health Sciences ,CAMP PRODUCTION ,3D invasion model ,Molecular Biology ,Arachidonic acid metabolite ,Pharmacology ,ECM ,Mesenchymal stem cell ,Biology and Life Sciences ,MYLK ,IN-VITRO ,ARACHIDONIC-ACID ,Cell Biology ,ENDOTHELIAL-CELLS ,Cell biology ,030104 developmental biology ,BARRIER FUNCTION ,030220 oncology & carcinogenesis ,LIGHT-CHAIN KINASE ,Molecular Medicine ,PHOSPHOLIPASE-C-EPSILON ,Intracellular - Abstract
Retraction of mesenchymal stromal cells supports the invasion of colorectal cancer cells (CRC) into the adjacent compartment. CRC-secreted 12(S)-HETE enhances the retraction of cancer-associated fibroblasts (CAFs) and therefore, 12(S)-HETE may enforce invasivity of CRC. Understanding the mechanisms of metastatic CRC is crucial for successful intervention. Therefore, we studied pro-invasive contributions of stromal cells in physiologically relevant three-dimensional in vitro assays consisting of CRC spheroids, CAFs, extracellular matrix and endothelial cells, as well as in reductionist models. In order to elucidate how CAFs support CRC invasion, tumour spheroid-induced CAF retraction and free intracellular Ca2+ levels were measured and pharmacological-or siRNA-based inhibition of selected signalling cascades was performed. CRC spheroids caused the retraction of CAFs, generating entry gates in the adjacent surrogate stroma. The responsible trigger factor 12(S)-HETE provoked a signal, which was transduced by PLC, IP3, free intracellular Ca2+, Ca(2+)calmodulin-kinase-II, RHO/ROCK and MYLK which led to the activation of myosin light chain 2, and subsequent CAF mobility. RHO activity was observed downstream as well as upstream of Ca2+ release. Thus, Ca2+ signalling served as central signal amplifier. Treatment with the FDA-approved drugs carbamazepine, cinnarizine, nifedipine and bepridil HCl, which reportedly interfere with cellular calcium availability, inhibited CAF-retraction. The elucidation of signalling pathways and identification of approved inhibitory drugs warrant development of intervention strategies targeting tumour-stroma interaction.
- Published
- 2016
46. Phosphorylation of NFATC1 at PIM1 target sites is essential for its ability to promote prostate cancer cell migration and invasion
- Author
-
Sini K, Eerola, Niina M, Santio, Sanni, Rinne, Petri, Kouvonen, Garry L, Corthals, Mauro, Scaravilli, Giovanni, Scala, Angela, Serra, Dario, Greco, Pekka, Ruusuvuori, Leena, Latonen, Eeva-Marja, Rainio, Tapio, Visakorpi, Päivi J, Koskinen, Institute of Biotechnology, Research Programs Unit, University Management, University of Helsinki, Eerola, Sini K., Santio, Niina M., Rinne, Sanni, Kouvonen, Petri, Corthals, Garry L., Scaravilli, Mauro, Scala, Giovanni, Serra, Angela, Greco, Dario, Ruusuvuori, Pekka, Latonen, Leena, Rainio, Eeva-Marja, Visakorpi, Tapio, Koskinen, Päivi J., Tampere University, BioMediTech, TAYS Cancer Centre, and Department of Clinical Chemistry
- Subjects
Male ,Metastatic carcinoma ,EXPRESSION ,NF-ATC ,3122 Cancers ,lcsh:Medicine ,Cell motility ,KAPPA-B ,ADHESION ,Mass Spectrometry ,ACTIVATION ,Proto-Oncogene Proteins c-pim-1 ,Cell Movement ,Tumor Cells, Cultured ,KINASE ,Humans ,Phosphorylation ,lcsh:QH573-671 ,Cell Proliferation ,Prostate cancer ,NFATC Transcription Factors ,integumentary system ,lcsh:Cytology ,Research ,lcsh:R ,PIM kinases ,PROLIFERATION ,Prostatic Neoplasms ,NFATC1 ,217 Medical engineering ,3126 Surgery, anesthesiology, intensive care, radiology ,TRANSCRIPTION FACTORS ,PC-3 Cells ,SURVIVAL ,1182 Biochemistry, cell and molecular biology ,OVEREXPRESSION ,Signal Transduction - Abstract
Background: Progression of prostate cancer from benign local tumors to metastatic carcinomas is a multistep process. Here we have investigated the signaling pathways that support migration and invasion of prostate cancer cells, focusing on the role of the NFATC1 transcription factor and its post-translational modifications. We have previously identified NFATC1 as a substrate for the PIM1 kinase and shown that PIM1-dependent phosphorylation increases NFATC1 activity without affecting its subcellular localization. Both PIM kinases and NFATC1 have been reported to promote cancer cell migration, invasion and angiogenesis, but it has remained unclear whether the effects of NFATC1 are phosphorylation-dependent and which downstream targets are involved. Methods: We used mass spectrometry to identify PIM1 phosphorylation target sites in NFATC1, and analysed their functional roles in three prostate cancer cell lines by comparing phosphodeficient mutants to wild-type NFATC1. We used luciferase assays to determine effects of phosphorylation on NFAT-dependent transcriptional activity, and migration and invasion assays to evaluate effects on cell motility. We also performed a microarray analysis to identify novel PIM1/NFATC1 targets, and validated one of them with both cellular expression analyses and in silico in clinical prostate cancer data sets. Results: Here we have identified ten PIM1 target sites in NFATC1 and found that prevention of their phosphorylation significantly decreases the transcriptional activity as well as the pro-migratory and pro-invasive effects of NFATC1 in prostate cancer cells. We observed that also PIM2 and PIM3 can phosphorylate NFATC1, and identified several novel putative PIM1/NFATC1 target genes. These include the ITGA5 integrin, which is differentially expressed in the presence of wild-type versus phosphorylation-deficient NFATC1, and which is coexpressed with PIM1 and NFATC1 in clinical prostate cancer specimens. Conclusions: Based on our data, phosphorylation of PIM1 target sites stimulates NFATC1 activity and enhances its ability to promote prostate cancer cell migration and invasion. Therefore, inhibition of the interplay between PIM kinases and NFATC1 may have therapeutic implications for patients with metastatic forms of cancer. publishedVersion
- Published
- 2019
47. Stat2 loss disrupts damage signalling and is protective in acute pancreatitis
- Author
-
Robert D. Goldin, Hiromi Kudo, Graham R. Foster, Gary Britton, Malcolm Ward, R. Hutchins, George Renney, William Alazawi, and H Heath
- Subjects
0301 basic medicine ,INCREASES ,medicine.medical_treatment ,ACTIVATION ,Mice ,0302 clinical medicine ,Interferon ,Pathology ,signalling ,Phosphorylation ,Mice, Knockout ,Chemistry ,MAP Kinase Kinase Kinases ,medicine.anatomical_structure ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,Acute Disease ,Acute pancreatitis ,PDX1 ,Cytokines ,Tumor necrosis factor alpha ,medicine.symptom ,Pancreas ,Life Sciences & Biomedicine ,Ceruletide ,medicine.drug ,Signal Transduction ,medicine.medical_specialty ,Inflammation ,KAPPA-B ,Arginine ,Pathology and Forensic Medicine ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Science & Technology ,business.industry ,Tumor Necrosis Factor-alpha ,1103 Clinical Sciences ,STAT2 Transcription Factor ,medicine.disease ,030104 developmental biology ,Endocrinology ,Pancreatitis ,Bone marrow ,business - Abstract
Severity of sterile inflammation, as seen in acute pancreatitis, is determined by damage-sensing receptors, signalling cascades and cytokine production. Stat2 is a type I interferon signalling mediator that also has interferon-independent roles in murine lipopolysaccharide-induced NF-κB-mediated sepsis. However its role in sterile inflammation is unknown. We hypothesised that Stat2 determines severity of non-infective inflammation in the pancreas.Wild type (WT) and Stat2−/− mice were injected intraperitoneally with cerulein or L-arginine. Specific cytokine-blocking antibodies were used in some experiments. Pancreata and blood were harvested 1h and 24h after the final dose of cerulein and up to 96h post L-arginine. Whole-tissue phosphoproteomic changes were assessed using label-free mass spectrometry. Tissue-specific Stat2 effects were studied in WT/Stat2−/− bone-marrow chimera and using Cre-lox recombination to delete Stat2 in pancreatic and duodenal homeobox 1(Pdx1)-expressing cells.Stat2−/− mice were protected from cerulein- and L-arginine-induced pancreatitis. Protection was independent of type I interferon signalling. Stat2−/− mice had lower cytokine levels including TNFα and IL-10 and reduced NF-kB nuclear localisation in pancreatic tissue compared to WT. Inhibition of TNFα improved (inhibition of IL-10 worsened) cerulein-induced pancreatitis in WT but not Stat2−/− mice. Phosphoproteomics showed down-regulation of mitogen-activated protein kinase (MAPK) mediators but accumulation of Ser412-phosphorylated Tak1. Stat2 deletion in Pdx1-expressing acinar cells (Stat2flox/Pdx1-cre) reduced pancreatic TNFα expression, but not histological injury or serum amylase. WT/Stat2−/− bone-marrow chimera were protected from pancreatitis irrespective of host or recipient genotype.Stat2 loss results in disrupted signalling in pancreatitis, upstream of NF-κB in non-acinar and/or bone marrow derived cells.
- Published
- 2019
48. A2ML1 and otitis media : novel variants, differential expression, and relevant pathways
- Author
-
Rachel Ann P Santos, Melquiadesa Pedro, Talitha Karisse L. Yarza, Charlotte M. Chiong, Janak A. Patel, Christopher Greenlee, Zubair M. Ahmed, Rose Anne Q Rosanes, Rehan S. Shaikh, Melissa A. Scholes, Matthew J. Steritz, Norman R. Friedman, Todd Wine, Abner L. Chan, Ma. Leah C. Tantoco, Patricia J. Yoon, Tori Bootpetch Roberts, Erasmo Gonzalo D V Llanes, Jeremy D. Prager, Anushree Acharya, Eric D. Larson, Karen L. Mohlke, Saima Riazuddin, Maria Rina T. Reyes-Quintos, Jose Pedrito M. Magno, Generoso T. Abes, Tasnee Chonmaitree, Petri S. Mattila, Teresa Luisa G. Cruz, Lena Hafrén, Elisabet Einarsdottir, Ayesha Yousaf, Catherine B. Anderson, Juha Kere, Jonathan Cardwell, Amanda G. Ruiz, Michael J. Bamshad, Herman A. Jenkins, Ivana V. Yang, Deborah A. Nickerson, Samuel P. Gubbels, Sven-Olrik Streubel, Suzanne M. Leal, Katerina Kechris, David A. Schwartz, Sheryl Mae Lagrana-Villagracia, Regie Lyn P. Santos-Cortez, Aileen Trinidad R Santos, Nanette R. Lee, Kenny H. Chan, Dylan Ray, Eva Maria Cutiongco-de la Paz, Stephen P. Cass, University Management, Research Programme of Molecular Medicine, Päivi Marjaana Saavalainen / Principal Investigator, Research Programs Unit, University of Helsinki, STEMM - Stem Cells and Metabolism Research Program, Juha Kere / Principal Investigator, Research Programme for Molecular Neurology, HUS Head and Neck Center, Korva-, nenä- ja kurkkutautien klinikka, Department of Ophthalmology and Otorhinolaryngology, and Clinicum
- Subjects
Male ,Proband ,DOWN-REGULATION ,A2ML1 ,Philippines ,SUSCEPTIBILITY ,Gene duplication ,Pakistan ,Child ,Exome ,Finland ,Genetics (clinical) ,Exome sequencing ,GENE-EXPRESSION ,Genetics ,Sanger sequencing ,0303 health sciences ,education.field_of_study ,030305 genetics & heredity ,1184 Genetics, developmental biology, physiology ,EPITHELIAL-CELLS ,Middle Aged ,Pedigree ,3. Good health ,READ ALIGNMENT ,TRANSCRIPTOME SIGNATURE ,Child, Preschool ,symbols ,Female ,medicine.symptom ,Signal Transduction ,Adult ,YOUNG-CHILDREN ,Adolescent ,Population ,RNA-sequencing ,KAPPA-B ,Biology ,INFLUENZA-A VIRUS ,Article ,Young Adult ,03 medical and health sciences ,symbols.namesake ,medicine ,Humans ,Genetic Predisposition to Disease ,alpha-Macroglobulins ,education ,030304 developmental biology ,alpha-2-macroglobulin-like-1 ,Sequence Analysis, RNA ,Gene Expression Profiling ,HEARING-LOSS ,Infant ,otitis media ,Sequence Analysis, DNA ,United States ,Otitis ,Gene Expression Regulation ,Mutation ,exome sequencing - Abstract
A genetic basis for otitis media is established, however, the role of rare variants in disease etiology is largely unknown. Previously a duplication variant within A2ML1 was identified as a significant risk factor for otitis media in an indigenous Filipino population and in US children. In this report exome and Sanger sequencing was performed using DNA samples from the indigenous Filipino population, Filipino cochlear implantees, US probands, Finnish, and Pakistani families with otitis media. Sixteen novel, damaging A2ML1 variants identified in otitis media patients were rare or low-frequency in population-matched controls. In the indigenous population, both gingivitis and A2ML1 variants including the known duplication variant and the novel splice variant c.4061 + 1 G>C were independently associated with otitis media. Sequencing of salivary RNA samples from indigenous Filipinos demonstrated lower A2ML1 expression according to the carriage of A2ML1 variants. Sequencing of additional salivary RNA samples from US patients with otitis media revealed differentially expressed genes that are highly correlated with A2ML1 expression levels. In particular, RND3 is upregulated in both A2ML1 variant carriers and high-A2ML1 expressors. These findings support a role for A2ML1 in keratinocyte differentiation within the middle ear as part of otitis media pathology and the potential application of ROCK inhibition in otitis media.
- Published
- 2019
49. Therapeutic glucocorticoids prevent bone loss but drive muscle wasting when administered in chronic polyarthritis
- Author
-
C. G. Fenton, J. M. Webster, C. S. Martin, S. Fareed, C. Wehmeyer, H. Mackie, R. Jones, A. P. Seabright, J. W. Lewis, Y. C. Lai, C. S. Goodyear, S. W Jones, M. S. Cooper, G. G. Lavery, R. Langen, K. Raza, R. S. Hardy, RS: NUTRIM - R3 - Respiratory & Age-related Health, and Pulmonologie
- Subjects
RISK ,lcsh:Diseases of the musculoskeletal system ,KAPPA-B ,OSTEOPOROSIS ,Muscle wasting ,RECOMMENDATIONS ,RHEUMATOID-ARTHRITIS ,PREVALENCE ,VERTEBRAL DEFORMITIES ,Polyarthritis ,FRACTURES ,MINERAL DENSITY ,lcsh:RC925-935 ,Glucocorticoids ,TUMOR-NECROSIS-FACTOR - Abstract
Background Patients with rheumatoid arthritis (RA) experience extra-articular manifestations including osteoporosis and muscle wasting, which closely associate with severity of disease. Whilst therapeutic glucocorticoids (GCs) reduce inflammation in RA, their actions on muscle and bone metabolism in the context of chronic inflammation remain unclear. We utilised the TNF-tg model of chronic polyarthritis to ascertain the impact of therapeutic GCs on bone and muscle homeostasis in the context of systemic inflammation. Methods TNF-tg and wild-type (WT) animals received either vehicle or the GC corticosterone (100 μg/ml) in drinking water at onset of arthritis. Arthritis severity and clinical parameters were measured, serum collected for ELISA and muscle and bone biopsies collected for μCT, histology and mRNA analysis. In vivo findings were examined in primary cultures of osteoblasts, osteoclasts and myotubes. Results TNF-tg mice receiving GCs showed protection from inflammatory bone loss, characterised by a reduction in serum markers of bone resorption, osteoclast numbers and osteoclast activity. In contrast, muscle wasting was markedly increased in WT and TNF-tg animals receiving GCs, independently of inflammation. This was characterised by a reduction in muscle weight and fibre size, and an induction in anti-anabolic and catabolic signalling. Conclusions This study demonstrates that when given in early onset chronic polyarthritis, oral GCs partially protect against inflammatory bone loss, but induce marked muscle wasting. These results suggest that in patients with inflammatory arthritis receiving GCs, the development of interventions to manage deleterious side effects in muscle should be prioritised.
- Published
- 2019
50. Disease-Associated SNPs in Inflammation-Related lncRNAs
- Author
-
Ainara Castellanos-Rubio and Sankar Ghosh
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Immunology ,kappa-b ,SNP ,Genome-wide association study ,Single-nucleotide polymorphism ,Computational biology ,Disease ,Review ,Biology ,Genome ,Polymorphism, Single Nucleotide ,intestinal-mucosa ,03 medical and health sciences ,0302 clinical medicine ,lncRNA ,Intestinal mucosa ,Immunology and Allergy ,Humans ,GWAS ,expression analysis ,Gene ,risk variants ,anril ,gene-expression ,Long non-coding RNA ,3. Good health ,inflammatory disease ,030104 developmental biology ,Gene Expression Regulation ,Immune System Diseases ,inflammation ,linc RNA ,genome-wide association ,rheumatoid-arthritis ,RNA, Long Noncoding ,long noncoding rna ,lcsh:RC581-607 ,celiac-disease ,030215 immunology ,Genome-Wide Association Study - Abstract
Immune-mediated diseases, such as celiac disease, type 1 diabetes or multiple sclerosis, are a clinically heterogeneous group of diseases that share many key genetic triggers. Although the pathogenic mechanisms responsible for the development of immune mediated disorders is not totally understood, high-throughput genomic studies, such as GWAS and Immunochip, performed in the past few years have provided intriguing hints about underlying mechanisms and pathways that lead to disease. More than a hundred gene variants associated with disease susceptibility have been identified through such studies, but the progress toward understanding the underlying mechanisms has been slow. The majority of the identified risk variants are located in non-coding regions of the genome making it difficult to assign a molecular function to the SNPs. However, recent studies have revealed that many of the non-coding regions bearing disease-associated SNPs generate long non-coding RNAs (lncRNAs). LncRNAs have been implicated in several inflammatory diseases, and many of them have been shown to function as regulators of gene expression. Many of the disease associated SNPs located in lncRNAs modify their secondary structure, or influence expression levels, thereby affecting their regulatory function, hence contributing to the development of disease. AC-R is funded by an Ikerbasque Research Fellowship, SG is funded by grant R01 DK102180 (NIH).
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.