12 research outputs found

    Analysis of Human TAAR8 and Murine Taar8b Mediated Signaling Pathways and Expression Profile

    Get PDF
    The thyroid hormone derivative 3-iodothyronamine (3-T1AM) exerts metabolic effects in vivo that contradict known effects of thyroid hormones. 3-T1AM acts as a trace amine-associated receptor 1 (TAAR1) agonist and activates Gs signaling in vitro. Interestingly, 3-T1AM-meditated in vivo effects persist in Taar1 knockout-mice indicating that further targets of 3-T1AM might exist. Here, we investigated another member of the TAAR family, the only scarcely studied mouse and human trace-amine-associated receptor 8 (Taar8b, TAAR8). By RT-qPCR and locked-nucleic-acid (LNA) in situ hybridization, Taar8b expression in different mouse tissues was analyzed. Functionally, we characterized TAAR8 and Taar8b with regard to cell surface expression and signaling via different G-protein-mediated pathways. Cell surface expression was verified by ELISA, and cAMP accumulation was quantified by AlphaScreen for detection of Gs and/or Gi/o signaling. Activation of G-proteins Gq/11 and G12/13 was analyzed by reporter gene assays. Expression analyses revealed at most marginal Taar8b expression and no gender differences for almost all analyzed tissues. In heart, LNA-in situ hybridization demonstrated the absence of Taar8b expression. We could not identify 3-T1AM as a ligand for TAAR8 and Taar8b, but both receptors were characterized by a basal Gi/o signaling activity, a so far unknown signaling pathway for TAARs

    Plasma proteome and metabolome characterization of an experimental human thyrotoxicosis model.

    Get PDF
    BACKGROUND: Determinations of thyrotropin (TSH) and free thyroxine (FT4) represent the gold standard in evaluation of thyroid function. To screen for novel peripheral biomarkers of thyroid function and to characterize FT4-associated physiological signatures in human plasma we used an untargeted OMICS approach in a thyrotoxicosis model. METHODS: A sample of 16 healthy young men were treated with levothyroxine for 8 weeks and plasma was sampled before the intake was started as well as at two points during treatment and after its completion, respectively. Mass spectrometry-derived metabolite and protein levels were related to FT4 serum concentrations using mixed-effect linear regression models in a robust setting. To compile a molecular signature discriminating between thyrotoxicosis and euthyroidism, a random forest was trained and validated in a two-stage cross-validation procedure. RESULTS: Despite the absence of obvious clinical symptoms, mass spectrometry analyses detected 65 metabolites and 63 proteins exhibiting significant associations with serum FT4. A subset of 15 molecules allowed a robust and good prediction of thyroid hormone function (AUC = 0.86) without prior information on TSH or FT4. Main FT4-associated signatures indicated increased resting energy expenditure, augmented defense against systemic oxidative stress, decreased lipoprotein particle levels, and increased levels of complement system proteins and coagulation factors. Further association findings question the reliability of kidney function assessment under hyperthyroid conditions and suggest a link between hyperthyroidism and cardiovascular diseases via increased dimethylarginine levels. CONCLUSION: Our results emphasize the power of untargeted OMICs approaches to detect novel pathways of thyroid hormone action. Furthermore, beyond TSH and FT4, we demonstrated the potential of such analyses to identify new molecular signatures for diagnosis and treatment of thyroid disorders. This study was registered at the German Clinical Trials Register (DRKS) [DRKS00011275] on the 16th of November 2016

    Integrierte Omics-Analysen zur Charakterisierung physiologischer Effekte von SchilddrĂĽsenhormonen und von spezifischen SchilddrĂĽsenhormon-Metaboliten

    No full text
    Die klassischen Schilddrüsenhormone (TH) Triiodthyronin (T3) und Thyroxin (T4) sind für die Regulation zahlreicher Stoffwechselprozesse von Bedeutung. Dabei beeinflussen sie unter anderem maßgeblich den hepatischen Energie- und Lipidstoffwechsel. In den letzten Jahren haben die beiden Schilddrüsenhormon-Metaboliten 3-Iodthyronamin (3-T1AM) und 3,5-Diiodthyronin (3,5-T2) an Aufmerksamkeit gewonnen, da sie in diversen Studien als endogene, biologisch aktive Substanzen beschrieben wurden. Die durch 3-T1AM-vermittelten metabolischen Effekte sind dabei denen der klassischen TH teilweise entgegengesetzt. Zudem konnte eine Interferenz mit der Hypothalamus-Hypophysen-Schilddrüsen (HPT)-Achse demonstriert werden. In dieser Arbeit sollte die Hypothese einer direkten 3-T1AM-Wirkung auf die Schilddrüse überprüft werden. Dazu wurden in einem in vitro-Modell 3-T1AM-behandelte Thyreozyten der Zelllinie PCCL3 mittels Transkriptomanalysen untersucht. Für den TH-Metaboliten 3,5-T2 konnten in früheren Arbeiten anti-steatotische, anti-lipidemische und kalorigene Effekte demonstriert werden. Aufgrund fehlender thyreotoxischer Nebenwirkungen, wie sie für die klassischen TH typisch sind, liegt ein therapeutisches Potenzial von 3,5-T2 zur Behandlung der mit steigender Inzidenz auftretenden Adipositas und der damit assoziierten Fettleber (Steatosis hepatis) nahe. Im Rahmen dieser Arbeit wurden die 3,5-T2-vermittelten Effekte auf die hepatischen Transkriptions- und Proteinmuster von Mäusen unter Standard- (SD) und Hochfettdiät (HFD) in komplementären Analysen charakterisiert. Die TH-Homöostase wird durch den negativen Rückkopplungsmechanismus der HPT-Achse reguliert. Im klinischen Alltag wird jedoch häufig eine Störung dieses Gleichgewichts in Form einer Hypo- oder Hyperthyreose beobachtet. Um die physiologischen Auswirkungen dieser Erkrankungen zu untersuchen, wurden in dieser Arbeit Proteomanalysen der Lebern von Mäusen mit induzierter Hypo- und Hyperthyreose durchgeführt, um bereits vorliegende korrespondierende Transkriptomdaten zu ergänzen. In den Transkriptomanalysen der 3-T1AM-behandelten Thyreozyten konnten keine Genexpressionsänderungen nachgewiesen werden. Um diese Diskrepanz zu den in anderen Studien demonstrierten metabolischen Effekten zu beheben, könnte eine Optimierung des experimentellen Designs sinnvoll sein. Alternativ könnte gefolgert werden, dass vor allem post-transkriptionelle Prozesse die Wirkungen von 3-T1AM vermitteln. Die komplementären Transkriptom- und Proteomdaten der 3,5-T2-behandelten Mäuse deuteten auf eine Stimulation der hepatischen Cholesterol-, Gallensäure- und lokalen Sexualhormon-Biosynthese in Tieren unter HFD hin. Außerdem konnten in Mäusen unter HFD erhöhte hepatische Spiegel von Sexualhormonen nachgewiesen werden. Weiterhin zeigten zahlreiche Transkripte und Proteine, welche in den Lipidstoffwechsel und Citratzyklus involviert sind, signifikante Mengenveränderungen nach 3,5-T2-Behandlung unter SD und HFD. Die in dieser Arbeit unter beiden Diäten beobachteten 3,5-T2-vermittelten Effekte auf Xenobiotika-metabolisierende Proteine könnten dabei unter anderem auf unerwünschte thyreomimetische Nebeneffekte hindeuten. Daher sollte der therapeutische Einsatz von 3,5-T2 als ein potenzielles anti-steatotisches Agens, wie es diverse vorangegangene Studien propagiert haben, kritisch betrachtet werden. Die ersten Ergebnisse der hepatischen Proteomanalysen hyperthyreoter Mäuse deuteten auf eine Reduktion von oxidativem Stress und eine Induktion der Proteinbiosynthese hin, während unter hypothyreoten Bedingungen entgegengesetzte Effekte beobachtet wurden. Im Rahmen dieser Arbeit konnten umfangreiche globale und komplementäre Datensätze mit Hilfe der Omics-Technologien Transkriptomics und Proteomics, für die Microarray- und Massenspektrometrie-basierte Analysen zum Einsatz kamen, generiert werden. Diese ermöglichten die Gewinnung neuer Erkenntnisse über die physiologischen Effekte und Wirkungsweisen der TH-Metabolite 3-T1AM und 3,5-T2 sowie die Krankheitsbilder Hypo- und Hyperthyreose.The classical thyroid hormones (TH) triiodothyronine (T3) and thyroxine (T4) regulate a variety of metabolic processes. Especially the hepatic energy and lipid metabolism is influenced by TH. In the last few years the TH metabolites 3-iodothyronamine (3-T1AM) and 3,5-diiodothyronine (3,5-T2) gained attention as they were demonstrated to represent endogenous biologically active compounds. The metabolic effects mediated by 3-T1AM are partially opposing to those of classical TH. Interference with the hypothalamic-pituitary-thyroid (HPT) axis could be demonstrated. The hypothesis of a direct action of 3-T1AM on the thyroid gland should be investigated in this thesis. Therefore, 3-T1AM-treated PCCL3 thyrocytes were analyzed in an in vitro model using transcriptome profiling. For the TH metabolite 3,5-T2 anti-steatotic, anti-lipidemic and calorigenic effects were demonstrated in previous studies. Due to obviously missing thyrotoxic side effects as they are typically observed for classical TH, 3,5-T2 was suggested as a potentially therapeutic agent against obesity and fatty liver (steatosis hepatis), which emerge with increasing incidence. In the context of this thesis 3,5-T2-mediated effects on hepatic transcriptome and proteome profiles from mice under standard (SD) and high-fat diet (HFD) were characterized in complementary analyses. The TH homeostasis is maintained by negative feedback regulation of the HPT axis. However, an imbalance of TH resulting in hypo- or hyperthyroidism is often observed in clinical daily routine. To analyze physiological consequences of these disorders, proteome analyses of livers from mice with induced hypo- and hyperthyroidism were performed, complementing previously generated corresponding transcriptome data. The transcriptome analyses of 3-T1AM-treated thyrocytes revealed no gene expression alterations. Since other studies demonstrated metabolic effects of 3-T1AM, this discrepancy might be due to different experimental designs. Alternatively, one could conclude that particularly post-transcriptional processes mediate 3-T1AM effects. Data of complementary transcriptome and proteome analyses from 3,5-T2-treated mice indicated stimulation of hepatic cholesterol, bile acid and local sex hormone biosynthesis in animals under HFD. Additionally, elevated hepatic levels of sex hormones were detected in mice under HFD. Furthermore, numerous transcripts and proteins involved in lipid metabolism and citric acid cycle exhibited significantly altered amounts after 3,5-T2 treatment under SD and HFD. The 3,5-T2-mediated effects on xenobiotic metabolizing proteins observed in these analyses under both diets could indicate adverse thyromimetic side effects. Thus, the therapeutic use of 3,5-T2 as a potentially anti-steatotic agent, as proposed by previous studies, should be assessed critically. The first results of the hepatic proteome analyses from hyperthyroid mice indicated reduced oxidative stress and induced protein biosynthesis, while under hypothyroid conditions opposing effects were observed. In the context of this thesis comprehensive global and complementary datasets were generated using the omics technologies transcriptomics and proteomics, where microarray- and mass spectrometry-based analyses were performed. These allowed to obtain new insights into physiological effects and modes of action of the TH metabolites 3-T1AM and 3,5-T2 as well as of the disorders hypo- and hyperthyroidism

    Overexpression of Transcripts Coding for Renin-b but Not for Renin-a Reduce Oxidative Stress and Increase Cardiomyoblast Survival under Starvation Conditions

    No full text
    A stimulated renin-angiotensin system is known to promote oxidative stress, apoptosis, necrosis and fibrosis. Renin transcripts (renin-b; renin-c) encoding a cytosolic renin isoform have been discovered that may in contrast to the commonly known secretory renin (renin-a) exert protective effects Here, we analyzed the effect of renin-a and renin-b overexpression in H9c2 cardiomyoblasts on apoptosis and necrosis as well as on potential mechanisms involved in cell death processes. To mimic ischemic conditions, cells were exposed to glucose starvation, anoxia or combined oxygen–glucose deprivation (OGD) for 24 h. Under OGD, control cells exhibited markedly increased necrotic and apoptotic cell death accompanied by enhanced ROS accumulation, loss of mitochondrial membrane potential and decreased ATP levels. The effects of OGD on necrosis were exaggerated in renin-a cells, but markedly diminished in renin-b cells. However, with respect to apoptosis, the effects of OGD were almost completely abolished in renin-b cells but interestingly also moderately diminished in renin-a cells. Under glucose depletion we found opposing responses between renin-a and renin-b cells; while the rate of necrosis and apoptosis was aggravated in renin-a cells, it was attenuated in renin-b cells. Based on our results, strategies targeting the regulation of cytosolic renin-b as well as the identification of pathways involved in the protective effects of renin-b may be helpful to improve the treatment of ischemia-relevant diseases

    Overexpression of Renin-B Induces Warburg-like Effects That Are Associated with Increased AKT/mTOR Signaling

    No full text
    The classical secretory renin-a is known to be involved in angiotensin generation, thereby regulating not only blood pressure, but also promoting oxidative stress as well as apoptotic and necrotic cell death. In contrast, another cytosolic renin isoform named renin-b has been described, exerting protective effects under ischemia-related conditions in H9c2 cardiomyoblasts. Using microarray-based transcriptome analyses, we aimed to identify the signaling pathways involved in mediating cardioprotection in H9c2 cells overexpressing renin-b. By transcriptome profiling, we identified increased gene expression of several genes encoding glycolytic enzymes and glucose transporters, while the transcript levels of TCA-cycle enzymes were decreased. Complementing data from metabolic analyses revealed enhanced glucose consumption and lactate accumulation due to renin-b overexpression. Renin-b overexpression further stimulated AKT/mTOR signaling, where numerous genes involved in this pathway showed altered transcript levels. For AKT, we also detected enhanced phosphorylation levels by means of Western blotting, suggesting an activation of this kinase. Moreover, analysis of the ROS levels identified an increase in ROS accumulation in renin-b-overexpressing cells. Altogether, our data demonstrate that renin-b overexpression induces the metabolic remodeling of H9c2 cells similar to that seen under oxygen deprivation. This metabolic phenotype exerting so-called aerobic glycolysis is also known as the Warburg effect

    B cells acquire a unique and differential transcriptomic profile during pregnancy

    No full text
    Pregnancy alters B cell development and function. B cell activation is initiated by antigens binding to the BCR leading to B cell survival, proliferation, antigen presentation and antibody production. We performed a genome-wide transcriptome profiling of splenic B cells from pregnant (P) and non-pregnant (NP) mice and identified 1136 genes exhibiting differential expression in B cells from P mice (625 up- and 511 down-regulated) compared to NP animals. In silico analysis showed that B cell activation through BCR seems to be lowered during pregnancy. RT-qPCR analysis confirmed these data. Additionally, B cells from pregnant women stimulated in vitro through BCR produced lower levels of inflammatory cytokines compared to non-pregnant women. Our results suggest that B cells acquire a state of hypo-responsiveness during gestation, probably as part of the maternal immune strategy for fetal tolerance but also open new avenues to understand why pregnant women are at highest risk for infections.Fil: Valeff, Natalin Jimena. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; ArgentinaFil: Muzzio, Damián Oscar. Greifswald University; AlemaniaFil: Matzner, Franziska. Greifswald University; AlemaniaFil: Dibo, Marcos Javier. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; ArgentinaFil: Golchert, Janine. University Medicine Greifswald; AlemaniaFil: Homuth, Georg. University Medicine Greifswald; AlemaniaFil: Abba, Martín Carlos. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. Universidad Nacional de La Plata. Facultad de Ciencias Médicas. Centro de Investigaciones Inmunológicas Básicas y Aplicadas; ArgentinaFil: Zygmunt, Marek. Greifswald University; AlemaniaFil: Jensen, Cristian Federico. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay. Centro de Estudios Farmacológicos y Botánicos. Universidad de Buenos Aires. Facultad de Medicina. Centro de Estudios Farmacológicos y Botánicos; Argentina. Universidad Bernardo O'Higgins; Chil

    DNA Polymerase Theta Plays a Critical Role in Pancreatic Cancer Development and Metastasis

    No full text
    Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly cancers worldwide. The occurrence of oncogenic KRAS mutations is considered a signature event in PDAC, leading to genomic instability. The aim of our study was to evaluate the impact of the oncogenic KRAS G12D mutation on the activity of the error-prone alt-EJ repair mechanism, and to investigate the potential role of Polθ in the development of pancreatic cancer. We found that oncogenic KRAS increases the expression of key alt-EJ proteins in a mouse and human PDAC model. Using TLR assay, we also found increased alt-EJ activity in mouse and human cell lines upon the expression of KRAS D12D. The inactivation/impairment of alt-EJ by polymerase theta (Polθ) depletion delays the development of pancreatic cancer and prolongs the survival of experimental mice, though it does not prevent the PDAC development, which leads to full-blown PDAC with disseminated metastasis. Our studies provide a high-value target as a novel therapeutic candidate for the treatment of pancreatic and other cancers. Abstract Pancreatic ductal adenocarcinoma (PDAC), due to its genomic heterogeneity and lack of effective treatment, despite decades of intensive research, will become the second leading cause of cancer-related deaths by 2030. Step-wise acquisition of mutations, due to genomic instability, is considered to drive the development of PDAC; the KRAS mutation occurs in 95 to 100% of human PDAC, and is already detectable in early premalignant lesions designated as pancreatic intraepithelial neoplasia (PanIN). This mutation is possibly the key event leading to genomic instability and PDAC development. Our study aimed to investigate the role of the error-prone DNA double-strand breaks (DSBs) repair pathway, alt-EJ, in the presence of the KRAS G12D mutation in pancreatic cancer development. Our findings show that oncogenic KRAS contributes to increasing the expression of Polθ, Lig3, and Mre11, key components of alt-EJ in both mouse and human PDAC models. We further confirm increased catalytic activity of alt-EJ in a mouse and human model of PDAC bearing the KRAS G12D mutation. Subsequently, we focused on estimating the impact of alt-EJ inactivation by polymerase theta (Polθ) deletion on pancreatic cancer development, and survival in genetically engineered mouse models (GEMMs) and cancer patients. Here, we show that even though Polθ deficiency does not fully prevent the development of pancreatic cancer, it significantly delays the onset of PanIN formation, prolongs the overall survival of experimental mice, and correlates with the overall survival of pancreatic cancer patients in the TCGA database. Our study clearly demonstrates the role of alt-EJ in the development of PDAC, and alt-EJ may be an attractive therapeutic target for pancreatic cancer patients

    In mouse chronic pancreatitis CD25+FOXP3+ regulatory T cells control pancreatic fibrosis by suppression of the type 2 immune response

    No full text
    Chronic pancreatitis (CP) is characterized by chronic inflammation and the progressive fibrotic replacement of exocrine and endocrine pancreatic tissue. We identify Treg cells as central regulators of the fibroinflammatory reaction by a selective depletion of FOXP3-positive cells in a transgenic mouse model (DEREG-mice) of experimental CP. In Treg-depleted DEREG-mice, the induction of CP results in a significantly increased stroma deposition, the development of exocrine insufficiency and significant weight loss starting from day 14 after disease onset. In CP, FOXP3+CD25+ Treg cells suppress the type-2 immune response by a repression of GATA3+ T helper cells (Th2), GATA3+ innate lymphoid cells type 2 (ILC2) and CD206+ M2-macrophages. A suspected pathomechanism behind the fibrotic tissue replacement may involve an observed dysbalance of Activin A expression in macrophages and of its counter regulator follistatin. Our study identified Treg cells as key regulators of the type-2 immune response and of organ remodeling during CP. The Treg/Th2 axis could be a therapeutic target to prevent fibrosis and preserve functional pancreatic tissue
    corecore