398 results on '"Thue W. Schwartz"'
Search Results
2. An atlas of GPCRs in dopamine neurons: Identification of the free fatty acid receptor 4 as a regulator of food and water intake
- Author
-
Mia Apuschkin, Hayley B. Burm, Jan H. Schmidt, Louise J. Skov, Rita C. Andersen, Carl-Fredrik Bowin, Jonatan F. Støier, Kathrine L. Jensen, Leonie P. Posselt, Oksana Dmytriyeva, Andreas T. Sørensen, Kristoffer L. Egerod, Birgitte Holst, Mattias Rickhag, Thue W. Schwartz, and Ulrik Gether
- Subjects
CP: Neuroscience ,CP: Metabolism ,Biology (General) ,QH301-705.5 - Abstract
Summary: Midbrain dopaminergic neurons (DANs) are subject to extensive metabotropic regulation, but the repertoire of G protein-coupled receptors (GPCRs) present in these neurons has not been mapped. Here, we isolate DANs from Dat-eGFP mice to generate a GPCR atlas by unbiased qPCR array expression analysis of 377 GPCRs. Combined with data mining of scRNA-seq databases, we identify multiple receptors in DAN subpopulations with 38 of these receptors representing the majority of transcripts. We identify 41 receptors expressed in midbrain DANs but not in non-DAN midbrain cells, including the free fatty acid receptor 4 (FFAR4). Functional expression of FFAR4 is validated by ex vivo Ca2+ imaging, and in vivo experiments support that FFAR4 negatively regulates food and water intake and bodyweight. In addition to providing a critical framework for understanding metabotropic DAN regulation, our data suggest fatty acid sensing by FFAR4 as a mechanism linking high-energy intake to the dopamine-reward pathway.
- Published
- 2024
- Full Text
- View/download PDF
3. Lactate receptor GPR81 drives breast cancer growth and invasiveness through regulation of ECM properties and Notch ligand DLL4
- Author
-
Kathrine Lundø, Oksana Dmytriyeva, Louise Spøhr, Eliana Goncalves-Alves, Jiayi Yao, Laia P. Blasco, Mette Trauelsen, Muthulakshmi Ponniah, Marc Severin, Albin Sandelin, Marie Kveiborg, Thue W. Schwartz, and Stine F. Pedersen
- Subjects
HCAR1 ,Tumor microenvironment ,Spheroid ,EPHA7 ,PCDH7 ,Notch ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The lactate receptor GPR81 contributes to cancer development through unclear mechanisms. Here, we investigate the roles of GPR81 in three-dimensional (3D) and in vivo growth of breast cancer cells and study the molecular mechanisms involved. Methods GPR81 was stably knocked down (KD) in MCF-7 human breast cancer cells which were subjected to RNA-seq analysis, 3D growth, in situ- and immunofluorescence analyses, and cell viability- and motility assays, combined with KD of key GPR81-regulated genes. Key findings were additionally studied in other breast cancer cell lines and in mammary epithelial cells. Results GPR81 was upregulated in multiple human cancer types and further upregulated by extracellular lactate and 3D growth in breast cancer spheroids. GPR81 KD increased spheroid necrosis, reduced invasion and in vivo tumor growth, and altered expression of genes related to GO/KEGG terms extracellular matrix, cell adhesion, and Notch signaling. Single cell in situ analysis of MCF-7 cells revealed that several GPR81-regulated genes were upregulated in the same cell clusters. Notch signaling, particularly the Notch ligand Delta-like-4 (DLL4), was strikingly downregulated upon GPR81 KD, and DLL4 KD elicited spheroid necrosis and inhibited invasion in a manner similar to GPR81 KD. Conclusions GPR81 supports breast cancer aggressiveness, and in MCF-7 cells, this occurs at least in part via DLL4. Our findings reveal a new GPR81-driven mechanism in breast cancer and substantiate GPR81 as a promising treatment target.
- Published
- 2023
- Full Text
- View/download PDF
4. Free fatty acid receptor 1 stimulates cAMP production and gut hormone secretion through Gq-mediated activation of adenylate cyclase 2
- Author
-
Jacob Emil Petersen, Maria Hauge Pedersen, Oksana Dmytriyeva, Emilie Nellemose, Tulika Arora, Maja Storm Engelstoft, Wesley B. Asher, Jonathan A. Javitch, Thue W. Schwartz, and Mette Trauelsen
- Subjects
FFAR1 ,GPR40 ,Adenylate cyclase 2 ,GLP-1 ,ADCY2 ,Gq ,Internal medicine ,RC31-1245 - Abstract
Objective: Free fatty acid receptor 1 (FFAR1) is highly expressed in enteroendocrine cells of the small intestine and pancreatic beta cells, where FFAR1 agonists function as GLP-1 and insulin secretagogues, respectively. Most efficacious are so-called second-generation synthetic agonists such as AM5262, which, in contrast to endogenous long-chain fatty acids are able to signal through both IP3/Ca2+ and cAMP pathways. Whereas IP3 signaling is to be expected for the mainly Gq-coupled FFAR1, the mechanism behind FFAR1-induced cAMP accumulation remains unclear, although originally proposed to be Gs mediated. Methods and results: When stimulated with AM5262, we observe that FFAR1 can activate the majority of the Gα proteins, except - surprisingly - members of the Gs family. AM5262-induced FFAR1-mediated transcriptional activation through cAMP response element (CREB) was blocked by the specific Gq inhibitor, YM253890. Furthermore, in Gq-deficient cells no CREB signal was observed unless Gq or G11 was reintroduced by transfection. By qPCR we determined that adenylate cyclase 2 (Adcy2) was highly expressed and enriched relative to the nine other Adcys in pro-glucagon expressing enteroendocrine cells. Co-transfection with ADCY2 increased the FFAR1-induced cAMP response 4-5-fold in WT HEK293 cells, an effect fully inhibited by YM253890. Moreover, co-transfection with ADCY2 had no effect in Gq-deficient cells without reintroduction of either Gq or G11. Importantly, although both AM5262/FFAR1 and isoproterenol/β2 adrenergic receptor (β2AR) induced cAMP production was lost in Gs-deficient cells, only the β2AR response was rescued by Gs transfection, whereas co-transfection with ADCY2 was required to rescue the FFAR1 cAMP response. In situ hybridization demonstrated a high degree of co-expression of ADCY2 and FFAR1 in enteroendocrine cells throughout the intestine. Finally, in the enteroendocrine STC-1 and GLUTag cell lines AM5262-induced cAMP accumulation and GLP-1 secretion were both blocked by YM253890. Conclusions: Our results show that Gq signaling is responsible not only for the IP3/Ca2+ but also the cAMP response, which together are required for the highly efficacious hormone secretion induced by second-generation FFAR1 agonists - and that ADCY2 presumably mediates the Gq-driven cAMP response.
- Published
- 2023
- Full Text
- View/download PDF
5. G-Protein-Coupled Receptor 91-Dependent Signalling Does Not Influence Vascular Inflammation and Atherosclerosis in Hyperlipidaemic Mice
- Author
-
Silke Griepke, Mette Trauelsen, Michelle D. Nilsson, Jakob Hansen, Lasse B. Steffensen, Thue W. Schwartz, and Daniel F. J. Ketelhuth
- Subjects
atherosclerosis ,inflammation ,GPR91 ,SUCNR1 ,succinate ,TCA ,Cytology ,QH573-671 - Abstract
The TCA cycle intermediate metabolite ‘succinate’ has been proposed as an inflammatory mediator, influencing autoimmunity and allergic reactions, through ligation to its sensing receptor SUCNR1/GPR91. Whether GPR91-mediated signalling influences the chronic inflammatory process of atherosclerosis has never been investigated. The examination of publicly available datasets revealed that the SUCNR1 gene is expressed in human atherosclerotic plaques, especially in vascular smooth muscle cells. Using GPR91 knockout (Gpr91−/−) and wildtype (WT) littermates, made hyperlipidaemic with the overexpression of the gain-of-function mutated Pcsk9 and Western diet feeding, we showed that the full ablation of GPR91 did not accelerate atherosclerosis—lesions in the aortic arch 2.18 ± 0.48% vs. 1.64 ± 0.31%, and in the aortic roots 10.06 ± 0.91% vs. 10.67 ± 1.53% for Gpr91−/− and WT mice, respectively. In line with this, no differences between groups were observed for macrophage and T-cell infiltration in the plaque, as well as the polarization towards M1- or M2-like macrophages in the aorta, spleen and liver of Gpr91−/− and WT control mice. In conclusion, our study indicates that the global ablation of GPR91 signalling does not influence vascular inflammation or atherogenesis.
- Published
- 2023
- Full Text
- View/download PDF
6. Self-organized metabotyping of obese individuals identifies clusters responding differently to bariatric surgery
- Author
-
Dimitra Lappa, Abraham S. Meijnikman, Kimberly A. Krautkramer, Lisa M. Olsson, Ömrüm Aydin, Anne-Sophie Van Rijswijk, Yair I. Z. Acherman, Maurits L. De Brauw, Valentina Tremaroli, Louise E. Olofsson, Annika Lundqvist, Siv A. Hjorth, Boyang Ji, Victor E. A. Gerdes, Albert K. Groen, Thue W. Schwartz, Max Nieuwdorp, Fredrik Bäckhed, and Jens Nielsen
- Subjects
Medicine ,Science - Abstract
Weight loss through bariatric surgery is efficient for treatment or prevention of obesity related diseases such as type 2 diabetes and cardiovascular disease. Long term weight loss response does, however, vary among patients undergoing surgery. Thus, it is difficult to identify predictive markers while most obese individuals have one or more comorbidities. To overcome such challenges, an in-depth multiple omics analyses including fasting peripheral plasma metabolome, fecal metagenome as well as liver, jejunum, and adipose tissue transcriptome were performed for 106 individuals undergoing bariatric surgery. Machine leaning was applied to explore the metabolic differences in individuals and evaluate if metabolism-based patients’ stratification is related to their weight loss responses to bariatric surgery. Using Self-Organizing Maps (SOMs) to analyze the plasma metabolome, we identified five distinct metabotypes, which were differentially enriched for KEGG pathways related to immune functions, fatty acid metabolism, protein-signaling, and obesity pathogenesis. The gut metagenome of the most heavily medicated metabotypes, treated simultaneously for multiple cardiometabolic comorbidities, was significantly enriched in Prevotella and Lactobacillus species. This unbiased stratification into SOM-defined metabotypes identified signatures for each metabolic phenotype and we found that the different metabotypes respond differently to bariatric surgery in terms of weight loss after 12 months. An integrative framework that utilizes SOMs and omics integration was developed for stratifying a heterogeneous bariatric surgery cohort. The multiple omics datasets described in this study reveal that the metabotypes are characterized by a concrete metabolic status and different responses in weight loss and adipose tissue reduction over time. Our study thus opens a path to enable patient stratification and hereby allow for improved clinical treatments.
- Published
- 2023
7. A systems biology approach to study non-alcoholic fatty liver (NAFL) in women with obesity
- Author
-
Abraham S. Meijnikman, Dimitra Lappa, Hilde Herrema, Omrum Aydin, Kimberly A. Krautkramer, Valentina Tremaroli, Louise E. Olofsson, Annika Lundqvist, Sjoerd Bruin, Yair Acherman, Joanne Verheij, Siv Hjorth, Victor E.A. Gerdes, Thue W. Schwartz, Albert K. Groen, Fredrik Bäckhed, Jens Nielsen, and Max Nieuwdorp
- Subjects
Biological sciences ,Physiology ,Human metabolism ,Systems biology ,Science - Abstract
Summary: Non-alcoholic fatty liver disease (NAFLD) is now the most frequent global chronic liver disease. Individuals with NAFLD exhibited an increased risk of all-cause mortality driven by extrahepatic cancers and liver and cardiovascular disease. Once the disease is established, women have a higher risk of disease progression and worse outcome. It is therefore critical to deepen the current knowledge on the pathophysiology of NAFLD in women. Here, we used a systems biology approach to investigate the contribution of different organs to this disease. We analyzed transcriptomics profiles of liver and adipose tissues, fecal metagenomes, and plasma metabolomes of 55 women with and without NAFLD. We observed differences in metabolites, expression of human genes, and gut microbial features between the groups and revealed that there is substantial crosstalk between these different omics sets. Multi-omics analysis of individuals with NAFLD may provide novel strategies to study the pathophysiology of NAFLD in humans.
- Published
- 2022
- Full Text
- View/download PDF
8. Adhesion receptor ADGRG2/GPR64 is in the GI-tract selectively expressed in mature intestinal tuft cells
- Author
-
Kaare V. Grunddal, Sarah Tonack, Kristoffer L. Egerod, Jonathan James Thompson, Natalia Petersen, Maja S. Engelstoft, Constance Vagne, Céline Keime, Gérard Gradwohl, Stefan Offermanns, and Thue W. Schwartz
- Subjects
ADGRG2 ,GPR64 ,GPCRs ,Tuft cells ,Chemosensory cells ,Internal medicine ,RC31-1245 - Abstract
Objective: GPR64/ADGRG2 is an orphan Adhesion G protein-coupled receptor (ADGR) known to be mainly expressed in the parathyroid gland and epididymis. This investigation aimed to delineate the cellular expression of GPR64 throughout the body with focus on the gastrointestinal (GI) tract. Methods: Transgenic Gpr64mCherry reporter mice were histologically examined throughout the body and reporter protein expression in intestinal tuft cells was confirmed by specific cell ablation. The GPCR repertoire of intestinal Gpr64mCherry-positive tuft cells was analyzed by quantitative RT-PCR analysis and in situ hybridization. The Gpr64mCherry was crossed into the general tuft cell reporter Trpm5GFP to generate small intestinal organoids for time-lapse imaging. Intestinal tuft cells were isolated from small intestine, FACS-purified and transcriptionally compared using RNA-seq analysis. Results: Expression of the Gpr64mCherry reporter was identified in multiple organs and specifically in olfactory microvillous cells, enteric nerves, and importantly in respiratory and GI tuft cells. In the small intestine, cell ablation targeting Gpr64-expressing epithelial cells eliminated tuft cells. Transcriptional analysis of small intestinal Gpr64mCherry -positive tuft cells confirmed expression of Gpr64 and the chemo-sensors Sucnr1, Gprc5c, Drd3, and Gpr41/Ffar3. Time-lapse studies of organoids from Trpm5GFP:Gpr64mCherry mice revealed sequential expression of initially Trpm5GFP and subsequently also Gpr64mCherry in maturing intestinal tuft cells. RNA-seq analysis of small intestinal tuft cells based on these two markers demonstrated a dynamic change in expression of transcription factors and GPCRs from young to mature tuft cells. Conclusions: GPR64 is expressed in chemosensory epithelial cells across a broad range of tissues; however, in the GI tract, GPR64 is remarkably selectively expressed in mature versus young immunoregulatory tuft cells.
- Published
- 2021
- Full Text
- View/download PDF
9. Extracellular succinate hyperpolarizes M2 macrophages through SUCNR1/GPR91-mediated Gq signaling
- Author
-
Mette Trauelsen, Thomas K. Hiron, Da Lin, Jacob E. Petersen, Billy Breton, Anna Sofie Husted, Siv A. Hjorth, Asuka Inoue, Thomas M. Frimurer, Michel Bouvier, Chris A. O’Callaghan, and Thue W. Schwartz
- Subjects
GPR91 ,SUCNR1 ,succinate ,M2 macrophages ,G protein ,Gq signaling ,Biology (General) ,QH301-705.5 - Abstract
Summary: Succinate functions both as a classical TCA cycle metabolite and an extracellular metabolic stress signal sensed by the mainly Gi-coupled succinate receptor SUCNR1. In the present study, we characterize and compare effects and signaling pathways activated by succinate and both classes of non-metabolite SUCNR1 agonists. By use of specific receptor and pathway inhibitors, rescue in G-protein-depleted cells and monitoring of receptor G protein activation by BRET, we identify Gq rather than Gi signaling to be responsible for SUCNR1-mediated effects on basic transcriptional regulation. Importantly, in primary human M2 macrophages, in which SUCNR1 is highly expressed, we demonstrate that physiological concentrations of extracellular succinate act through SUCNR1-activated Gq signaling to efficiently regulate transcription of immune function genes in a manner that hyperpolarizes their M2 versus M1 phenotype. Thus, sensing of stress-induced extracellular succinate by SUCNR1 is an important transcriptional regulator in human M2 macrophages through Gq signaling.
- Published
- 2021
- Full Text
- View/download PDF
10. Rfx6 promotes the differentiation of peptide-secreting enteroendocrine cells while repressing genetic programs controlling serotonin production
- Author
-
Julie Piccand, Constance Vagne, Florence Blot, Aline Meunier, Anthony Beucher, Perrine Strasser, Mari L. Lund, Sabitri Ghimire, Laure Nivlet, Céline Lapp, Natalia Petersen, Maja S. Engelstoft, Christelle Thibault-Carpentier, Céline Keime, Sara Jimenez Correa, Valérie Schreiber, Nacho Molina, Thue W. Schwartz, Adèle De Arcangelis, and Gérard Gradwohl
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: Enteroendocrine cells (EECs) of the gastro-intestinal tract sense gut luminal factors and release peptide hormones or serotonin (5-HT) to coordinate energy uptake and storage. Our goal is to decipher the gene regulatory networks controlling EECs specification from enteroendocrine progenitors. In this context, we studied the role of the transcription factor Rfx6 which had been identified as the cause of Mitchell–Riley syndrome, characterized by neonatal diabetes and congenital malabsorptive diarrhea. We previously reported that Rfx6 was essential for pancreatic beta cell development and function; however, the role of Rfx6 in EECs differentiation remained to be elucidated. Methods: We examined the molecular, cellular, and metabolic consequences of constitutive and conditional deletion of Rfx6 in the embryonic and adult mouse intestine. We performed single cell and bulk RNA-Seq to characterize EECs diversity and identify Rfx6-regulated genes. Results: Rfx6 is expressed in the gut endoderm; later, it is turned on in, and restricted to, enteroendocrine progenitors and persists in hormone-positive EECs. In the embryonic intestine, the constitutive lack of Rfx6 leads to gastric heterotopia, suggesting a role in the maintenance of intestinal identity. In the absence of intestinal Rfx6, EECs differentiation is severely impaired both in the embryo and adult. However, the number of serotonin-producing enterochromaffin cells and mucosal 5-HT content are increased. Concomitantly, Neurog3-positive enteroendocrine progenitors accumulate. Combined analysis of single-cell and bulk RNA-Seq data revealed that enteroendocrine progenitors differentiate in two main cell trajectories, the enterochromaffin (EC) cells and the Peptidergic Enteroendocrine (PE) cells, the differentiation programs of which are differentially regulated by Rfx6. Rfx6 operates upstream of Arx, Pax6 and Isl1 to trigger the differentiation of peptidergic EECs such as GIP-, GLP-1-, or CCK-secreting cells. On the contrary, Rfx6 represses Lmx1a and Tph1, two genes essential for serotonin biosynthesis. Finally, we identified transcriptional changes uncovering adaptive responses to the prolonged lack of enteroendocrine hormones and leading to malabsorption and lower food efficiency ratio in Rfx6-deficient mouse intestine. Conclusion: These studies identify Rfx6 as an essential transcriptional regulator of EECs specification and shed light on the molecular mechanisms of intestinal failures in human RFX6-deficiencies such as Mitchell–Riley syndrome. Keywords: Rfx6, Neurog3, Lmx1a, Intestine, Cell fate, Serotonin, Enteroendocrine cells, Enterochromaffin cells, Malabsorption, Mitchell–Riley syndrome
- Published
- 2019
- Full Text
- View/download PDF
11. Human substance P receptor binding mode of the antagonist drug aprepitant by NMR and crystallography
- Author
-
Shuanghong Chen, Mengjie Lu, Dongsheng Liu, Lingyun Yang, Cuiying Yi, Limin Ma, Hui Zhang, Qing Liu, Thomas M. Frimurer, Ming-Wei Wang, Thue W. Schwartz, Raymond C. Stevens, Beili Wu, Kurt Wüthrich, and Qiang Zhao
- Subjects
Science - Abstract
The FDA approved drug aprepitant is an antagonist of the Neurokinin 1 receptor (NK1R). Here the authors present aprepitant bound NK1R crystal structures and use NMR spectroscopy to gain further insights into the dynamics of aprepitant binding, which is of interest for further drug development.
- Published
- 2019
- Full Text
- View/download PDF
12. Autocrine negative feedback regulation of lipolysis through sensing of NEFAs by FFAR4/GPR120 in WAT
- Author
-
Anna Sofie Husted, Jeppe H. Ekberg, Emma Tripp, Tinne A.D. Nissen, Stijn Meijnikman, Shannon L. O'Brien, Trond Ulven, Yair Acherman, Sjoerd C. Bruin, Max Nieuwdorp, Zach Gerhart-Hines, Davide Calebiro, Lars O. Dragsted, and Thue W. Schwartz
- Subjects
GPR120 ,FFAR4 ,Autocrine ,Lipolysis ,NEFA ,GPCR ,Internal medicine ,RC31-1245 - Abstract
Objectives: Long-chain fatty acids (LCFAs) released from adipocytes inhibit lipolysis through an unclear mechanism. We hypothesized that the LCFA receptor, FFAR4 (GPR120), which is highly expressed in adipocytes, may be involved in this feedback regulation. Methods and results: Liquid chromatography mass spectrometry (LC-MS) analysis of conditioned media from isoproterenol-stimulated primary cultures of murine and human adipocytes demonstrated that most of the released non-esterified free fatty acids (NEFAs) are known agonists for FFAR4. In agreement with this, conditioned medium from isoproterenol-treated adipocytes stimulated signaling strongly in FFAR4 transfected COS-7 cells as opposed to non-transfected control cells. In transfected 3T3-L1 cells, FFAR4 agonism stimulated Gi- and Go-mini G protein binding more strongly than Gq, effects which were blocked by the selective FFAR4 antagonist AH7614. In primary cultures of murine white adipocytes, the synthetic, selective FFAR4 agonist CpdA inhibited isoproterenol-induced intracellular cAMP accumulation in a manner similar to the antilipolytic control agent nicotinic acid acting through another receptor, HCAR2. In vivo, oral gavage with the synthetic, specific FFAR4 agonist CpdB decreased the level of circulating NEFAs in fasting lean mice to a similar degree as nicotinic acid. In agreement with the identified anti-lipolytic effect of FFAR4, plasma NEFAs and glycerol were increased in FFAR4-deficient mice as compared to littermate controls despite having elevated insulin levels, and cAMP accumulation in primary adipocyte cultures was augmented by treatment with the FFAR4 antagonist conceivably by blocking the stimulatory tone of endogenous NEFAs on FFAR4. Conclusions: In white adipocytes, FFAR4 functions as an NEFA-activated, autocrine, negative feedback regulator of lipolysis by decreasing cAMP though Gi-mediated signaling.
- Published
- 2020
- Full Text
- View/download PDF
13. Profiling of G protein-coupled receptors in vagal afferents reveals novel gut-to-brain sensing mechanisms
- Author
-
Kristoffer L. Egerod, Natalia Petersen, Pascal N. Timshel, Jens C. Rekling, Yibing Wang, Qinghua Liu, Thue W. Schwartz, and Laurent Gautron
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objectives: G protein-coupled receptors (GPCRs) act as transmembrane molecular sensors of neurotransmitters, hormones, nutrients, and metabolites. Because unmyelinated vagal afferents richly innervate the gastrointestinal mucosa, gut-derived molecules may directly modulate the activity of vagal afferents through GPCRs. However, the types of GPCRs expressed in vagal afferents are largely unknown. Here, we determined the expression profile of all GPCRs expressed in vagal afferents of the mouse, with a special emphasis on those innervating the gastrointestinal tract. Methods: Using a combination of high-throughput quantitative PCR, RNA sequencing, and in situ hybridization, we systematically quantified GPCRs expressed in vagal unmyelinated Nav1.8-expressing afferents. Results: GPCRs for gut hormones that were the most enriched in Nav1.8-expressing vagal unmyelinated afferents included NTSR1, NPY2R, CCK1R, and to a lesser extent, GLP1R, but not GHSR and GIPR. Interestingly, both GLP1R and NPY2R were coexpressed with CCK1R. In contrast, NTSR1 was coexpressed with GPR65, a marker preferentially enriched in intestinal mucosal afferents. Only few microbiome-derived metabolite sensors such as GPR35 and, to a lesser extent, GPR119 and CaSR were identified in the Nav1.8-expressing vagal afferents. GPCRs involved in lipid sensing and inflammation (e.g. CB1R, CYSLTR2, PTGER4), and neurotransmitters signaling (CHRM4, DRD2, CRHR2) were also highly enriched in Nav1.8-expressing neurons. Finally, we identified 21 orphan GPCRs with unknown functions in vagal afferents. Conclusion: Overall, this study provides a comprehensive description of GPCR-dependent sensing mechanisms in vagal afferents, including novel coexpression patterns, and conceivably coaction of key receptors for gut-derived molecules involved in gut-brain communication. Keywords: G protein-coupled receptors, Vagal afferent nerves, Gut-brain axis, Gut hormones, GLP1R, NTSR1
- Published
- 2018
- Full Text
- View/download PDF
14. Enterochromaffin 5-HT cells – A major target for GLP-1 and gut microbial metabolites
- Author
-
Mari L. Lund, Kristoffer L. Egerod, Maja S. Engelstoft, Oksana Dmytriyeva, Elvar Theodorsson, Bhavik A. Patel, and Thue W. Schwartz
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objectives: 5-HT storing enterochromaffin (EC) cells are believed to respond to nutrient and gut microbial components, and 5-HT receptor-expressing afferent vagal neurons have been described to be the major sensors of nutrients in the GI-tract. However, the molecular mechanism through which EC cells sense nutrients and gut microbiota is still unclear. Methods and results: TPH1, the 5-HT generating enzyme, and chromogranin A, an acidic protein responsible for secretory granule storage of 5-HT, were highly enriched in FACS-purified EC cells from both small intestine and colon using a 5-HT antibody-based method. Surprisingly, EC cells from the small intestine did not express GPCR sensors for lipid and protein metabolites, such as FFAR1, GPR119, GPBAR1 (TGR5), CaSR, and GPR142, in contrast to the neighboring GLP-1 storing enteroendocrine cell. However, the GLP-1 receptor was particularly highly expressed and enriched in EC cells as judged both by qPCR and by immunohistochemistry using a receptor antibody. GLP-1 receptor agonists robustly stimulated 5-HT secretion from intestinal preparations using both HPLC and a specific amperometric method. Colonic EC cells expressed many different types of known and potential GPCR sensors of microbial metabolites including three receptors for SCFAs, i.e. FFAR2, OLF78, and OLF558 and receptors for aromatic acids, GPR35; secondary bile acids GPBAR1; and acyl-amides and lactate, GPR132. Conclusion: Nutrient metabolites apparently do not stimulate EC cells of the small intestine directly but through a paracrine mechanism involving GLP-1 secreted from neighboring enteroendocrine cells. In contrast, colonic EC cells are able to sense a multitude of different metabolites generated by the gut microbiota as well as gut hormones, including GLP-1. Keywords: Metabolite GPCR, Nutrient sensing, Gut microbiota, Gut hormone, Enteroendocrine, Afferent vagal nerves
- Published
- 2018
- Full Text
- View/download PDF
15. Receptor structure-based discovery of non-metabolite agonists for the succinate receptor GPR91
- Author
-
Mette Trauelsen, Elisabeth Rexen Ulven, Siv A. Hjorth, Matjaz Brvar, Claudia Monaco, Thomas M. Frimurer, and Thue W. Schwartz
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: Besides functioning as an intracellular metabolite, succinate acts as a stress-induced extracellular signal through activation of GPR91 (SUCNR1) for which we lack suitable pharmacological tools. Methods and results: Here we first determined that the cis conformation of the succinate backbone is preferred and that certain backbone modifications are allowed for GPR91 activation. Through receptor modeling over the X-ray structure of the closely related P2Y1 receptor, we discovered that the binding pocket is partly occupied by a segment of an extracellular loop and that succinate therefore binds in a very different mode than generally believed. Importantly, an empty side-pocket is identified next to the succinate binding site. All this information formed the basis for a substructure-based search query, which, combined with molecular docking, was used in virtual screening of the ZINC database to pick two serial mini-libraries of a total of only 245 compounds from which sub-micromolar, selective GPR91 agonists of unique structures were identified. The best compounds were backbone-modified succinate analogs in which an amide-linked hydrophobic moiety docked into the side-pocket next to succinate as shown by both loss- and gain-of-function mutagenesis. These compounds displayed GPR91-dependent activity in altering cytokine expression in human M2 macrophages similar to succinate, and importantly were devoid of any effect on the major intracellular target, succinate dehydrogenase. Conclusions: These novel, synthetic non-metabolite GPR91 agonists will be valuable both as pharmacological tools to delineate the GPR91-mediated functions of succinate and as leads for the development of GPR91-targeted drugs to potentially treat low grade metabolic inflammation and diabetic complications such as retinopathy and nephropathy. Keywords: GPR91, SUCNR1, Metabolite receptor, GPCR, Drug discovery, Virtual screening, Chemical design
- Published
- 2017
- Full Text
- View/download PDF
16. Disruption of GPR35 Signaling in Bone Marrow-Derived Cells Does Not Influence Vascular Inflammation and Atherosclerosis in Hyperlipidemic Mice
- Author
-
Roland Baumgartner, Felipe B. Casagrande, Randi B. Mikkelsen, Martin Berg, Konstantinos A. Polyzos, Maria J. Forteza, Aastha Arora, Thue W. Schwartz, Siv A. Hjorth, and Daniel F. J. Ketelhuth
- Subjects
atherosclerosis ,GPR35 ,inflammation ,immunometabolism ,kynurenine ,tryptophan ,Microbiology ,QR1-502 - Abstract
G-protein-coupled receptor-35 (GPR35) has been identified as a receptor for the tryptophan metabolite kynurenic acid (KynA) and suggested to modulate macrophage polarization in metabolic tissues. Whether GPR35 can influence vascular inflammation and atherosclerosis has however never been tested. Lethally irradiated LdlrKO mice were randomized to receive GPR35KO or wild type (WT) bone marrow transplants and fed a high cholesterol diet for eight weeks to develop atherosclerosis. GPR35KO and WT chimeric mice presented no difference in the size of atherosclerotic lesions in the aortic arch (2.37 ± 0.58% vs. 1.95 ± 0.46%, respectively) or in the aortic roots (14.77 ± 3.33% vs. 11.57 ± 2.49%, respectively). In line with these data, no changes in the percentage of VCAM-1+, IAb + cells, and CD3+ T cells, as well as alpha smooth muscle cell actin expression, was observed between groups. Interestingly, the GPR35KO group presented a small but significant increase in CD68+ macrophage infiltration in the plaque. However, in vitro culture experiments using bone marrow-derived macrophages from both groups indicated that GPR35 plays no role in modulating the secretion of major inflammatory cytokines. Our study indicates that GPR35 expression does not play a direct role in macrophage activation, vascular inflammation, and the development of atherosclerosis.
- Published
- 2021
- Full Text
- View/download PDF
17. β-Cell Specific Overexpression of GPR39 Protects against Streptozotocin-Induced Hyperglycemia
- Author
-
Kristoffer L. Egerod, Chunyu Jin, Pia Steen Petersen, Nils Wierup, Frank Sundler, Birgitte Holst, and Thue W. Schwartz
- Subjects
Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Mice deficient in the zinc-sensor GPR39, which has been demonstrated to protect cells against endoplasmatic stress and cell death in vitro, display moderate glucose intolerance and impaired glucose-induced insulin secretion. Here, we use the Tet-On system under the control of the proinsulin promoter to selectively overexpress GPR39 in the β cells in a double transgenic mouse strain and challenge them with multiple low doses of streptozotocin, which in the wild-type littermates leads to a gradual increase in nonfasting glucose levels and glucose intolerance observed during both food intake and OGTT. Although the overexpression of the constitutively active GPR39 receptor in animals not treated with streptozotocin appeared by itself to impair the glucose tolerance slightly and to decrease the β-cell mass, it nevertheless totally protected against the gradual hyperglycemia in the steptozotocin-treated animals. It is concluded that GPR39 functions in a β-cell protective manner and it is suggested that it is involved in some of the beneficial, β-cell protective effects observed for Zn++ and that GPR39 may be a target for antidiabetic drug intervention.
- Published
- 2011
- Full Text
- View/download PDF
18. Cell selectivity in succinate receptor SUCNR1/GPR91 signaling in skeletal muscle
- Author
-
Ahmed M. Abdelmoez, Oksana Dmytriyeva, Yasemin-Xiomara Zurke, Mette Trauelsen, Alesandra A. Marica, Mladen Savikj, Jonathon A. B. Smith, Claudia Monaco, Thue W. Schwartz, Anna Krook, and Nicolas J. Pillon
- Subjects
Physiology ,Physiology (medical) ,Endocrinology, Diabetes and Metabolism - Abstract
Macrophages but not skeletal muscle cells respond to extracellular succinate via SUCNR1/GPR91.
- Published
- 2023
19. Hyperinsulinemia Is Highly Associated With Markers of Hepatocytic Senescence in Two Independent Cohorts
- Author
-
Abraham S. Meijnikman, Casper C. van Olden, Ömrüm Aydin, Hilde Herrema, Dorota Kaminska, Dimitra Lappa, Ville Männistö, Valentina Tremaroli, Louise E. Olofsson, Maurits de Brauw, Arnold van de Laar, Joanne Verheij, Victor E.A. Gerdes, Thue W. Schwartz, Jens Nielsen, Fredrik Bäckhed, Päivi Pajukanta, Jussi Pihlajamäki, Tamar Tchkonia, James L. Kirkland, Folkert Kuipers, Max Nieuwdorp, Albert K. Groen, Pathology, Internal medicine, ACS - Diabetes & metabolism, AGEM - Endocrinology, metabolism and nutrition, Center for Liver, Digestive and Metabolic Diseases (CLDM), Graduate School, Vascular Medicine, ACS - Atherosclerosis & ischemic syndromes, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, and Experimental Vascular Medicine
- Subjects
Diabetes Mellitus, Type 2 ,Liver ,Non-alcoholic Fatty Liver Disease ,Endocrinology, Diabetes and Metabolism ,Hyperinsulinism ,Internal Medicine ,Humans ,Insulin ,Insulin Resistance ,Biomarkers - Abstract
Cellular senescence is an essentially irreversible growth arrest that occurs in response to various cellular stressors and may contribute to development of type 2 diabetes and Non-Alcoholic Fatty Liver Disease (NAFLD). Here, we investigated whether chronically elevated insulin levels are associated with cellular senescence in the human liver. In 107 individuals undergoing bariatric surgery, hepatic senescence markers were assessed by immunohistochemistry as well as transcriptomics. A subset of 180 participants from the ongoing Finnish Kuopio OBesity Surgery (KOBS) study was used as validation cohort. We found plasma insulin to be highly associated with various markers of cellular senescence in liver tissue. The liver transcriptome of individuals with high insulin revealed significant upregulation of several genes associated with senescence: p21, TGFβ, PI3K, HLA-G, IL8, p38, Ras, and E2F. Insulin associated with hepatic senescence independently of NAFLD and plasma glucose. By using transcriptomic data from the KOBS study, we could validate the association of insulin with p21 in the liver. Our results support a potential role for hyperinsulinemia in induction of cellular senescence in the liver. These findings suggest possible benefits of lowering insulin levels in obese individuals with insulin resistance.
- Published
- 2022
20. Paracrine relationship between incretin hormones and endogenous 5‐hydroxytryptamine in the small and large intestine
- Author
-
Iain R. Tough, Mari L. Lund, Bhavik A. Patel, Thue W. Schwartz, and Helen M. Cox
- Subjects
Endocrine and Autonomic Systems ,Physiology ,Gastroenterology - Published
- 2023
21. Pyruvate dehydrogenase kinase regulates vascular inflammation in atherosclerosis and increases cardiovascular risk
- Author
-
Maria J Forteza, Martin Berg, Andreas Edsfeldt, Jangming Sun, Roland Baumgartner, Ilona Kareinen, Felipe Beccaria Casagrande, Ulf Hedin, Song Zhang, Ivan Vuckovic, Petras P Dzeja, Konstantinos A Polyzos, Anton Gisterå, Mette Trauelsen, Thue W Schwartz, Lea Dib, Joerg Herrmann, Claudia Monaco, Ljubica Matic, Isabel Gonçalves, and Daniel F J Ketelhuth
- Subjects
Physiology ,Physiology (medical) ,Cardiology and Cardiovascular Medicine - Abstract
AimsRecent studies have revealed a close connection between cellular metabolism and the chronic inflammatory process of atherosclerosis. While the link between systemic metabolism and atherosclerosis is well established, the implications of altered metabolism in the artery wall are less understood. Pyruvate dehydrogenase kinase (PDK)-dependent inhibition of pyruvate dehydrogenase (PDH) has been identified as a major metabolic step regulating inflammation. Whether the PDK/PDH axis plays a role in vascular inflammation and atherosclerotic cardiovascular disease remains unclear.Methods and resultsGene profiling of human atherosclerotic plaques revealed a strong correlation between PDK1 and PDK4 transcript levels and the expression of pro-inflammatory and destabilizing genes. Remarkably, the PDK1 and PDK4 expression correlated with a more vulnerable plaque phenotype, and PDK1 expression was found to predict future major adverse cardiovascular events. Using the small-molecule PDK inhibitor dichloroacetate (DCA) that restores arterial PDH activity, we demonstrated that the PDK/PDH axis is a major immunometabolic pathway, regulating immune cell polarization, plaque development, and fibrous cap formation in Apoe−/− mice. Surprisingly, we discovered that DCA regulates succinate release and mitigates its GPR91-dependent signals promoting NLRP3 inflammasome activation and IL-1β secretion by macrophages in the plaque.ConclusionsWe have demonstrated for the first time that the PDK/PDH axis is associated with vascular inflammation in humans and particularly that the PDK1 isozyme is associated with more severe disease and could predict secondary cardiovascular events. Moreover, we demonstrate that targeting the PDK/PDH axis with DCA skews the immune system, inhibits vascular inflammation and atherogenesis, and promotes plaque stability features in Apoe−/− mice. These results point toward a promising treatment to combat atherosclerosis.
- Published
- 2023
22. Activation of succinate receptor 1 boosts human mast cell reactivity and allergic bronchoconstriction
- Author
-
Xiao Tang, Elin Rönnberg, Jesper Säfholm, Madhuranayaki Thulasingam, Mette Trauelsen, Thue W. Schwartz, Craig E. Wheelock, Sven‐Erik Dahlén, Gunnar Nilsson, and Jesper Z. Haeggström
- Subjects
Inflammation ,Respiratory Medicine and Allergy ,Bronchoconstriction ,Guinea Pigs ,Immunology ,Succinates ,Immunoglobulin E ,succinate ,Asthma ,SUCNR1 ,eicosanoid ,Hypersensitivity ,Animals ,Humans ,Immunology and Allergy ,mast cell hyper-reactivity ,Mast Cells ,allergic bronchoconstriction ,Lungmedicin och allergi - Abstract
Background: SUCNR1 is a sensor of extracellular succinate, a Krebs cycle intermediate generated in excess during oxidative stress and has been linked to metabolic regulation and inflammation. While mast cells express SUCNR1, its role in mast cell reactivity and allergic conditions such as asthma remains to be elucidated. Methods: Cord blood-derived mast cells and human mast cell line LAD-2 challenged by SUCNR1 ligands were analyzed for the activation and mediator release. Effects on mast cell-dependent bronchoconstriction were assessed in guinea pig trachea and isolated human small bronchi challenged with antigen and anti-IgE, respectively. Results: SUCNR1 is abundantly expressed on human mast cells. Challenge with succinate, or the synthetic non-metabolite agonist cis-epoxysuccinate, renders mast cells hypersensitive to IgE-dependent activation, resulting in augmented degranulation and histamine release, de novo biosynthesis of eicosanoids and cytokine secretion. The succinate-potentiated mast cell reactivity was attenuated by SUCNR1 knockdown and selective SUCNR1 antagonists and could be tuned by pharmacologically targeting protein kinase C and extracellular signal-regulated kinase. Both succinate and cis-epoxysuccinate dose-dependently potentiated antigen-induced contraction in a mast cell-dependent guinea pig airway model, associated with increased generation of cysteinyl-leukotrienes and histamine in trachea. Similarly, cis-epoxysuccinate aggravated IgE-receptor-induced contraction of human bronchi, which was blocked by SUCNR1 antagonism. Conclusion: SUCNR1 amplifies IgE-receptor-induced mast cell activation and allergic bronchoconstriction, suggesting a role for this pathway in aggravation of allergic asthma, thus linking metabolic perturbations to mast cell-dependent inflammation.
- Published
- 2022
23. Molecular Dynamics Based Identification of Binding Pathways and Two Distinct High-Affinity Sites for Succinate in SUCNR1/GPR91
- Author
-
Aslihan Shenol, Michael Lückmann, Mette Trauelsen, Matteo Lambrughi, Matteo Tiberti, Elena Papaleo, Thomas Michael Frimurer, and Thue W. Schwartz
- Published
- 2023
24. Vagal afferent cholecystokinin receptor activation is required for glucagon‐like peptide‐1–induced satiation
- Author
-
Myrtha Arnold, Harald S. Hansen, Jens F. Rehfeld, Michelle K. Lærke, Thue W. Schwartz, Oksana Dmytriyeva, Wolfgang Langhans, and Vasiliki Vana
- Subjects
FFA1 receptor ,medicine.medical_specialty ,TAK875 ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Intraperitoneal injection ,Enteroendocrine cell ,vagus ,Satiation ,digestive system ,Cholecystokinin receptor ,Eating ,Mice ,Endocrinology ,Glucagon-Like Peptide 1 ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Humans ,Receptor ,Cholecystokinin ,business.industry ,CCK ,digestive, oral, and skin physiology ,Vagus Nerve ,Glucagon-like peptide-1 ,Vagus nerve ,Anorectic ,Receptors, Cholecystokinin ,GLP-1 ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
Peripheral glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK) are secreted from enteroendocrine cells, and their plasma concentrations increase in response to eating. While the satiating effect of gut-derived CCK on food intake control is well documented, the effect of peripheral GLP-1 is less clear. There is evidence that native GLP-1 can inhibit food intake only in the fed state but not in the fasting state. We therefore hypothesised that other gut peptides released during a meal might influence the subsequent effect of endogenous GLP-1 and investigated whether CCK could do so. We found that intraperitoneal injection of CCK in food-restricted mice inhibited food intake during the first 30 min segment of a one-hour session of ad libitum chow intake and that mice compensated by increasing their intake during the second half of the session. Importantly, this compensatory behaviour was abolished by an intraperitoneal injection of GLP-1 administered following an intraperitoneal injection of CCK and prior to the one-hour session. In vivo activation of the FFA1 receptor with orally administered TAK875 increased plasma CCK concentration and, consistent with the effect of exogenous CCK, we found that prior oral administration of TAK875 increased the eating inhibitory effect of peripherally administered GLP-1. To examine the role of the vagus nerve in this effect, we utilised a saporin-based lesioning procedure to selectively ablate the CCK receptor-expressing gastrointestinal vagal afferent neurones (VANs). We found that the combined anorectic effect of TAK875 and GLP-1 was significantly attenuated in the absence of CCK receptor expressing VANs. Taken together, our results indicate that endogenous CCK interacts with GLP-1 to promote satiation and that activation of the FFA1 receptor can initiate this interaction by stimulating the release of CCK. This article is protected by copyright. All rights reserved.
- Published
- 2021
25. Outrunning obesity with Lac-Phe?
- Author
-
Christoffer Clemmensen, Jens Lund, and Thue W. Schwartz
- Subjects
Physiology ,Phenylalanine ,Humans ,Cell Biology ,Lactic Acid ,Obesity ,Muscle, Skeletal ,Molecular Biology ,Exercise - Abstract
Lactate released from skeletal muscle during high-intensity exercise gives rise to a surge in circulating lactate-derived pseudo-dipeptide metabolites including N-lactoyl-phenylalanine (Lac-Phe). In a recent Nature paper, Li et al. use genetic and pharmacological evidence to now propose Lac-Phe to be an "exercise hormone" that suppresses appetite and obesity.
- Published
- 2022
26. Amino acids differ in their capacity to stimulate GLP-1 release from the perfused rat small intestine and stimulate secretion by different sensing mechanisms
- Author
-
Mette M. Rosenkilde, Stella Feng Sheng Xu, Sara L. Jepsen, Cathrine Ørskov, Rune E. Kuhre, Thue W. Schwartz, Ida M. Modvig, Maja S. Engelstoft, Jens J. Holst, and Kristoffer L. Egerod
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Physiology ,Endocrinology, Diabetes and Metabolism ,Mice, Transgenic ,030209 endocrinology & metabolism ,GPR3 ,Rat Small Intestine ,Receptors, G-Protein-Coupled ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Glucagon-Like Peptide 1 ,Physiology (medical) ,Internal medicine ,Intestine, Small ,medicine ,Animals ,Secretion ,Amino Acids ,Rats, Wistar ,Receptors, Lysophosphatidic Acid ,chemistry.chemical_classification ,Secretory Pathway ,Chemistry ,Glucagon-like peptide-1 ,Small intestine ,Rats ,Amino acid ,Mice, Inbred C57BL ,Perfusion ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Signal Transduction - Abstract
The aim of this study was to explore individual amino acid-stimulated GLP-1 responses and the underlying stimulatory mechanisms, as well as to identify the amino acid-sensing-receptors involved in amino acid-stimulated GLP-1 release. Experiments were primarily based on isolated perfused rat small intestines, which have intact epithelial polarization allowing discrimination between luminal and basolateral mechanisms as well as quantitative studies of intestinal absorption and hormone secretion. Expression analysis of amino acid sensors on isolated murine GLP-1 secreting L-cells was assessed by qPCR. We found that L-valine powerfully stimulated GLP-1 secretion but only from the luminal side (2.9-fold increase). When administered from the vascular side, L-arginine and the aromatic amino acids stimulated GLP-1 secretion equally (2.6-2.9 fold increases). Expression analysis revealed that Casr expression was enriched in murine GLP-1 secreting L-cells, whereas Gpr35, Gprc6a, Gpr142, Gpr93 (Lpar5) and the umami taste receptor subunits Tas1r3 and Tas1r1 were not. Consistently, activation of GPR35, GPR93, GPR142 and the umami taste receptor with specific agonists or allosteric modulators did not increase GLP-1 secretion (P>0.05 for all experiments), whereas vascular inhibition of CaSR reduced GLP-1 secretion in response to luminal infusion of mixed amino acids. In conclusion, amino acids differ in their capacity to stimulate GLP-1 secretion. Some amino acids stimulated secretion only from the intestinal lumen, while other amino acids exclusively stimulated secretion from the vascular side, indicating that amino acid-stimulated GLP-1 secretion involves both apical and basolateral (post-absorptive) sensing mechanisms. Sensing of absorbed amino acids involves CaSR activation as vascular inhibition of CaSR markedly diminished amino acid stimulated GLP-1 release.
- Published
- 2021
27. Systems analysis of metabolic responses to a mixed meal test in an obese cohort reveals links between tissue metabolism and the gut microbiota
- Author
-
Peishun Li, Boyang Ji, Dimitra Lappa, Abraham S Meijnikman, Lisa M. Olsson, Ömrüm Aydin, Sjoerd C. Bruin, Arnold van de Laar, Valentina Tremaroli, Hao Luo, Jun Geng, Kimberly A. Krautkramer, Annika Lundqvist, Hilde Herrema, Albert K. Groen, Victor E.A. Gerdes, Thue W. Schwartz, Fredrik Bäckhed, Max Nieuwdorp, Louise E. Olofsson, and Jens Nielsen
- Abstract
Individuals with prediabetes and type 2 diabetes mellitus (T2DM) have poor ability to adapt to diet-triggered perturbations. We investigated global metabolic responses to a mixed meal test (MMT) in morbidly obese individuals with different diabetic status by performing plasma metabolomic profiling. Abnormal metabolism of carbohydrates, (branched-chain) amino acids, fatty acids and acylcholines in individuals with (pre)diabetes was observed. Moreover, differences in metabolic responses were associated with altered fecal metagenomics and transcriptomes of liver, jejunum and adipose tissues, which revealed a modified gut microbiome and multi-tissue metabolism in individuals having insulin resistance. Finally, using integrative machine learning models, we built a predictive model based on metabolomics data after 2h MMT, and identified possible new biomarkers for glycemic control including N−acetylaspartate and phenylalanine-derived metabolites that may be useful for diagnosis, intervention and prevention of T2DM.
- Published
- 2022
28. Jan Fahrenkrug (6/6 1947 – 10/11 2021)
- Author
-
Jens F. Rehfeld, Jens Hannibal, Jens Juul Holst, Tomas Hökfelt, Ove B. Schaffalitzky de Muckadell, and Thue W. Schwartz
- Subjects
Cellular and Molecular Neuroscience ,Endocrinology ,Physiology ,Biochemistry - Published
- 2022
29. Protective succinate-SUCNR1 metabolic stress signaling gone bad
- Author
-
Thue W. Schwartz, Mette Trauelsen, and Sally Winther
- Subjects
Physiology ,Metabolite ,Inflammation ,Cell Biology ,Metabolism ,medicine.disease ,Phenotype ,Cell biology ,Citric acid cycle ,chemistry.chemical_compound ,chemistry ,Fibrosis ,Extracellular ,medicine ,medicine.symptom ,Receptor ,Molecular Biology - Abstract
The TCA cycle metabolite succinate functions as an intra- and extracellular signal of metabolic stress. Based on the phenotype of UCP-1-deficient mice, Mills et al. (2021) now report in Nature Metabolism that accumulation of extracellular succinate due to impaired elimination in thermogenic fat drives liver inflammation and fibrosis through the succinate receptor SUCNR1.
- Published
- 2021
30. snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis
- Author
-
Aviv Regev, Eugene Drokhlyansky, Jozef Ukropec, Christian Wolfrum, Lucia Balazova, Gottfried Rudofsky, Wenfei Sun, Antonio Giordano, Lianggong Ding, Miroslav Balaz, Anna Sofie Husted, Patrik Stefanicka, Georgia Colleluori, Michal Slyper, Saverio Cinti, Zuzana Kovanicova, Thue W. Schwartz, and Hua Dong
- Subjects
Adult ,Male ,0301 basic medicine ,genetic processes ,Paracrine Communication ,Regulator ,Adipose tissue ,Cell Separation ,Acetates ,Biology ,Aldehyde Dehydrogenase 1 Family ,Article ,Mice ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,Paracrine signalling ,0302 clinical medicine ,Adipose Tissue, Brown ,Single-cell analysis ,Adipocyte ,Adipocytes ,Animals ,Humans ,RNA-Seq ,Aged ,Cell Nucleus ,Multidisciplinary ,Retinal Dehydrogenase ,Cytochrome P-450 CYP2E1 ,Thermogenesis ,Middle Aged ,Cell biology ,030104 developmental biology ,chemistry ,Female ,Single-Cell Analysis ,Energy Metabolism ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Adipose tissue is usually classified on the basis of its function as white, brown or beige (brite)1. It is an important regulator of systemic metabolism, as shown by the fact that dysfunctional adipose tissue in obesity leads to a variety of secondary metabolic complications2,3. In addition, adipose tissue functions as a signalling hub that regulates systemic metabolism through paracrine and endocrine signals4. Here we use single-nucleus RNA-sequencing (snRNA-seq) analysis in mice and humans to characterize adipocyte heterogeneity. We identify a rare subpopulation of adipocytes in mice that increases in abundance at higher temperatures, and we show that this subpopulation regulates the activity of neighbouring adipocytes through acetate-mediated modulation of their thermogenic capacity. Human adipose tissue contains higher numbers of cells of this subpopulation, which could explain the lower thermogenic activity of human compared to mouse adipose tissue and suggests that targeting this pathway could be used to restore thermogenic activity.
- Published
- 2020
31. Evidence that a deviation in the kynurenine pathway aggravates atherosclerotic disease in humans
- Author
-
Thue W. Schwartz, Ulf Hedin, Roland Baumgartner, Daniel F. J. Ketelhuth, Siv A. Hjorth, Göran K. Hansson, Ljubica Perisic Matic, Gabrielle Paulsson-Berne, K P Polyzos, Martin Berg, and Maria J. Forteza
- Subjects
ARYL-HYDROCARBON RECEPTOR ,Carotid Artery Diseases ,0301 basic medicine ,Kynurenine pathway ,INHIBITION ,Regulator ,Down-Regulation ,Inflammation ,030204 cardiovascular system & hematology ,Pharmacology ,Kynurenic Acid ,CORONARY EVENTS ,IDO ,ACTIVATION ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Kynurenic acid ,Downregulation and upregulation ,kynurenic acid ,In vivo ,Internal Medicine ,medicine ,Humans ,Macrophage ,REGULATORY T-CELLS ,Kynurenine ,biology ,aryl hydrocarbon receptor ,business.industry ,CARDIOVASCULAR RISK ,Macrophages ,Tryptophan ,Aryl hydrocarbon receptor ,Plaque, Atherosclerotic ,TRYPTOPHAN ,Up-Regulation ,030104 developmental biology ,chemistry ,QUINOLINIC ACID ,inflammation ,biology.protein ,atherosclerosis ,medicine.symptom ,business ,INDOLEAMINE 2,3-DIOXYGENASE ,kynurenine pathway - Abstract
BACKGROUND: The metabolism of tryptophan (Trp) along the kynurenine pathway has been shown to carry strong immunoregulatory properties. Several experimental studies indicate that this pathway is a major regulator of vascular inflammation and influences atherogenesis. Knowledge of the role of this pathway in human atherosclerosis remains incomplete.OBJECTIVES: In this study, we performed a multiplatform analysis of tissue samples, in vitro and in vivo functional assays to elucidate the potential role of the kynurenine pathway in human atherosclerosis.METHODS AND RESULTS: Comparison of transcriptomic data from carotid plaques and control arteries revealed an upregulation of enzymes within the quinolinic branch of the kynurenine pathway in the disease state, whilst the branch leading to the formation of kynurenic acid (KynA) was downregulated. Further analyses indicated that local inflammatory responses are closely tied to the deviation of the kynurenine pathway in the vascular wall. Analysis of cerebrovascular symptomatic and asymptomatic carotid stenosis data showed that the downregulation of KynA branch enzymes and reduced KynA production were associated with an increased probability of patients to undergo surgery due to an unstable disease. In vitro, we showed that KynA-mediated signalling through aryl hydrocarbon receptor (AhR) is a major regulator of human macrophage activation. Using a mouse model of peritoneal inflammation, we showed that KynA inhibits leukocyte recruitment.CONCLUSIONS: We have found that a deviation in the kynurenine pathway is associated with an increased probability of developing symptomatic unstable atherosclerotic disease. Our study suggests that KynA-mediated signalling through AhR is an important mechanism involved in the regulation of vascular inflammation.
- Published
- 2020
32. Activation of metabolite receptor GPR91 promotes platelet aggregation and transcellular biosynthesis of leukotriene C4
- Author
-
Nailin Li, Mette Trauelsen, Xiao Tang, David Fuchs, Jesper Z. Haeggström, Shuai Tan, Thue W. Schwartz, and Craig E. Wheelock
- Subjects
biology ,Leukotriene C4 ,GPR91 ,Chemistry ,Thromboxane ,Hematology ,Lipid signaling ,030204 cardiovascular system & hematology ,succinate ,antiplatelet therapy ,eicosanoids ,Cell biology ,Proinflammatory cytokine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Phospholipase A2 ,platelet aggregation ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Platelet ,Arachidonic acid ,Platelet activation - Abstract
Background Succinate is a Krebs cycle intermediate whose formation is enhanced under metabolic stress, and for which a selective sensor GPR91 has been identified on various cell types including platelets. Platelet-derived eicosanoids play pivotal roles in platelet activation/aggregation, which is key to thrombus formation and progression of atherothrombosis. Objectives This study aims to decipher the molecular mechanism(s) and potential involvement of eicosanoids in succinate enhanced platelet activation/aggregation. Methods We used liquid chromatography-mass spectrometry (LC-MS)/MS-based lipid mediator profiling to identify eicosanoids regulated by succinate. We ran light transmittance aggregometry and flow cytometry to assess platelet aggregation, P-selectin expression, and platelet-polymorphonuclear leukocyte (PMN) adherence. Various pharmacological tools were used to assess the contributions of GPR91 signalling and eicosanoids in platelet aggregation. Results Succinate and two types of synthetic non-metabolite GPR91 agonists-cis-epoxysuccinate (cES) and Cmpd131-potentiated platelet aggregation, which was partially blocked by a selective GPR91 antagonist XT1. GPR91 activation increased production of 12-hydroxy-eicosatetraenoic acid (12-HETE), thromboxane (TX) A2 , and 12-hydroxy-heptadecatrienoic acid (12-HHT) in human platelets, associated with phosphorylation of cytosolic phospholipase A2 (cPLA2 ), suggesting increased availability of free arachidonic acid. Blocking 12-HETE and TXA2 synthesis, or antagonism of the TXA2 receptor, significantly reduced platelet aggregation enhanced by GPR91 signalling. Moreover, platelet-PMN suspensions challenged with succinate exhibited enhanced transcellular biosynthesis of leukotriene C4 (LTC4 ), a powerful proinflammatory vascular spasmogen. Conclusion Succinate signals through GPR91 to promote biosynthesis of eicosanoids, which contribute to platelet aggregation/activation and potentially vascular inflammation. Hence, GPR91 may be a suitable target for pharmacological intervention in atherothrombotic conditions.
- Published
- 2020
33. Optimization of First-in-Class Dual-Acting FFAR1/FFAR4 Allosteric Modulators with Novel Mode of Action
- Author
-
Michael Lückmann, Aslihan Shenol, Tinne A. D. Nissen, Jacob E. Petersen, David Kouvchinov, Thue W. Schwartz, and Thomas M. Frimurer
- Subjects
GPR40 ,FFAR4 ,G-protein-coupled receptor ,GPR120 ,Organic Chemistry ,Drug Discovery ,free fatty acid receptor ,Biochemistry ,FFAR1 - Abstract
The free fatty acid receptors FFAR1 and FFAR4 are considered promising therapeutic targets for management of metabolic and inflammatory diseases. However, there is a need for entirely novel chemical scaffolds, since many of the highly similar lipophilic chemotypes in development have been abandoned by the pharmaceutical industry, due to toxic effects on hepatocytes and β-cells. Our group has recently reported the discovery of a 1,3,5-triazine-2-amine-based compound that acts as an allosteric agonist on FFAR1. Here, we present the synthesis and investigation of the structure-activity relationship of an extensive set of analogues of which many display dual-acting agonist properties for both FFAR1 and FFAR4. In several rounds of optimization, we discovered multiple analogues with single-digit nanomolar potency on FFAR1. Pending additional optimization for metabolic stability, the compounds in this study present novel ways of providing beneficial glycemic control while avoiding the notorious toxicity challenges associated with previously identified chemotypes.
- Published
- 2022
34. Glucagon-like peptide 1 (GLP-1)
- Author
-
Kerstin Stemmer, Stephen C. Woods, Harvey J. Grill, Jens J. Holst, Alessandro Pocai, Michael A. Nauck, Andreas Fritsche, Richard D. DiMarchi, David A. D'Alessio, Mads Tang-Christensen, Diego Perez-Tilve, Darleen A. Sandoval, Randy J. Seeley, Wolfgang Langhans, Daniel J. Drucker, Stephen R. Bloom, M.H. Tschöp, Brian Finan, Fiona M. Gribble, Timo D. Müller, Peter R. Flatt, Joel F. Habener, Juris J. Meier, Thue W. Schwartz, Frank Reimann, Gribble, Fiona [0000-0002-4232-2898], Reimann, Frank [0000-0001-9399-6377], and Apollo - University of Cambridge Repository
- Subjects
Blood Glucose ,0301 basic medicine ,endocrine system ,lcsh:Internal medicine ,medicine.medical_treatment ,Incretin ,030209 endocrinology & metabolism ,Review ,Gastric Inhibitory Polypeptide ,Pharmacology ,Neuroprotection ,Glucagon-Like Peptide-1 Receptor ,03 medical and health sciences ,0302 clinical medicine ,GLP-1 ,Insulin ,Glucagon ,Diabetes ,Obesity ,Glucagon-Like Peptide 1 ,Insulin-Secreting Cells ,Diabetes mellitus ,Insulin Secretion ,Receptors, Glucagon ,medicine ,Humans ,Hypoglycemic Agents ,ddc:610 ,Receptor ,lcsh:RC31-1245 ,Molecular Biology ,2. Zero hunger ,Gastric emptying ,business.industry ,digestive, oral, and skin physiology ,Cell Biology ,medicine.disease ,Glucagon-like peptide-1 ,ddc ,3. Good health ,Glucose ,030104 developmental biology ,Diabetes Mellitus, Type 2 ,business ,hormones, hormone substitutes, and hormone antagonists ,Hormone - Abstract
Background: The glucagon-like peptide-1 (GLP-1) is a multifaceted hormone with broad pharmacological potential. Among the numerous metabolic effects of GLP-1 are the glucose-dependent stimulation of insulin secretion, decrease of gastric emptying, inhibition of food intake, increase of natriuresis and diuresis, and modulation of rodent beta-cell proliferation. GLP-1 also has cardio- and neuroprotective effects, decreases inflammation and apoptosis, and has implications for learning and memory, reward behavior, and palatability. Biochemically modified for enhanced potency and sustained action, GLP-1 receptor agonists are successfully in clinical use for the treatment of type-2 diabetes, and several GLP-1-based pharmacotherapies are in clinical evaluation for the treatment of obesity. Scope of review: In this review, we provide a detailed overview on the multifaceted nature of GLP-1 and its pharmacology and discuss its therapeutic implications on various diseases. Major conclusions: Since its discovery, GLP-1 has emerged as a pleiotropic hormone with a myriad of metabolic functions that go well beyond its classical identification as an incretin hormone. The numerous beneficial effects of GLP-1 render this hormone an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, and neurodegenerative disorders (C) 2019 The Authors. Published by Elsevier GmbH.
- Published
- 2019
35. Author response for 'Vagal afferent CCK receptor activation is required for GLP ‐1‐ induced satiation'
- Author
-
Jens F. Rehfeld, Vasiliki Vana, Myrtha Arnold, Thue W. Schwartz, Harald S. Hansen, Wolfgang Langhans, Michelle K. Lærke, and Oksana Dmytriyeva
- Subjects
medicine.medical_specialty ,Endocrinology ,business.industry ,Internal medicine ,medicine ,Vagal afferent ,business ,Cholecystokinin receptor - Published
- 2021
36. Metabolite G-protein coupled receptor signaling: Potential regulation of eicosanoids
- Author
-
Xiao, Tang, Yaolin, Hou, Thue W, Schwartz, and Jesper Z, Haeggström
- Subjects
Proteomics ,Pharmacology ,Arachidonic Acid ,Eicosanoids ,Lipid Metabolism ,Biochemistry ,Receptors, G-Protein-Coupled - Abstract
Eicosanoids are a family of bioactive compounds derived from arachidonic acid (AA) that play pivotal roles in physiology and disease, including inflammatory conditions of multiple organ systems. The biosynthesis of eicosanoids requires a series of catalytic steps that are controlled by designated enzymes, which can be regulated by inflammatory and stress signals via transcriptional and translational mechanisms. In the past decades, evidence have emerged indicating that G-protein coupled receptors (GPCRs) can sense extracellular metabolites, and regulate inflammatory responses including eicosanoid production. This review focuses on the recent advances of metabolite GPCRs research, their role in regulation of eicosanoid biosynthesis, and the link to pathophysiological conditions.
- Published
- 2022
37. L-Cell Differentiation Is Induced by Bile Acids Through GPBAR1 and Paracrine GLP-1 and Serotonin Signaling
- Author
-
Fredrik Bäckhed, Chantal Kroone, Thue W. Schwartz, Eelco J.P. de Koning, Natalia Petersen, Kristina Schoonjans, Giovanni Sorrentino, Filip K. Knop, Frank Reimann, Kristoffer L. Egerod, Fiona M. Gribble, Brynjulf Mortensen, Mari L. Lund, Daniel J. Drucker, Mari Lilith, Lund, Sorrentino, G, Kristoffer Lihme, Egerod, Chantal, Kroone, Brynjulf, Mortensen, Filip Krag, Knop, Frank, Reimann, Fiona, M Gribble, Daniel, J Drucker, Eelco JP de, Koning, Kristina, Schoonjan, Fredrik, Bäckhed, Thue, W Schwartz, Natalia, Petersen, Knop, Filip Krag [0000-0002-2495-5034], Gribble, Fiona M [0000-0002-4232-2898], Drucker, Daniel J [0000-0001-6688-8127], Petersen, Natalia [0000-0001-9344-6041], Apollo - University of Cambridge Repository, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
0301 basic medicine ,Male ,pyy ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,neurotensin ,Receptors, G-Protein-Coupled ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Glucagon-Like Peptide 1 ,Receptor ,organoids ,GUT MICROBIOTA ,EXPRESSION ,NEUROTENSIN ,MECHANISMS ,mechanisms ,digestive, oral, and skin physiology ,Cell Differentiation ,G protein-coupled bile acid receptor ,Cell biology ,gs ,Jejunum ,gq ,Female ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,Agonist ,endocrine system ,Serotonin ,Lithocholic acid ,medicine.drug_class ,Enteroendocrine Cells ,Paracrine Communication ,030209 endocrinology & metabolism ,Glucagon-Like Peptide-1 Receptor ,Article ,Bile Acids and Salts ,03 medical and health sciences ,Paracrine signalling ,expression ,Internal Medicine ,medicine ,Animals ,Humans ,mouse ,gut microbiota ,030104 developmental biology ,chemistry - Abstract
Glucagon-like peptide 1 (GLP-1) mimetics are effective drugs for treatment of type 2 diabetes, and there is consequently extensive interest in increasing endogenous GLP-1 secretion and L-cell abundance. Here we identify G-protein–coupled bile acid receptor 1 (GPBAR1) as a selective regulator of intestinal L-cell differentiation. Lithocholic acid and the synthetic GPBAR1 agonist, L3740, selectively increased L-cell density in mouse and human intestinal organoids and elevated GLP-1 secretory capacity. L3740 induced expression of Gcg and transcription factors Ngn3 and NeuroD1. L3740 also increased the L-cell number and GLP-1 levels and improved glucose tolerance in vivo. Further mechanistic examination revealed that the effect of L3740 on L cells required intact GLP-1 receptor and serotonin 5-hydroxytryptamine receptor 4 (5-HT4) signaling. Importantly, serotonin signaling through 5-HT4 mimicked the effects of L3740, acting downstream of GLP-1. Thus, GPBAR1 agonists and other powerful GLP-1 secretagogues facilitate L-cell differentiation through a paracrine GLP-1–dependent and serotonin-mediated mechanism.
- Published
- 2020
38. Disruption of GPR35 Signaling in Bone Marrow-Derived Cells Does Not Influence Vascular Inflammation and Atherosclerosis in Hyperlipidemic Mice
- Author
-
Maria J. Forteza, Martin Berg, Thue W. Schwartz, Siv A. Hjorth, Daniel F. J. Ketelhuth, Randi B. Mikkelsen, Konstantinos A. Polyzos, Felipe B. Casagrande, Roland Baumgartner, and Aastha Arora
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,immunometabolism ,Macrophage polarization ,Inflammation ,Microbiology ,Biochemistry ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Kynurenic acid ,Internal medicine ,medicine ,Macrophage ,tryptophan ,Receptor ,Molecular Biology ,CD68 ,QR1-502 ,kynurenine ,macrophages ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,inflammation ,Bone marrow ,medicine.symptom ,atherosclerosis ,KYNURENIC ACID ,030217 neurology & neurosurgery ,GPR35 - Abstract
G-protein-coupled receptor-35 (GPR35) has been identified as a receptor for the tryptophan metabolite kynurenic acid (KynA) and suggested to modulate macrophage polarization in metabolic tissues. Whether GPR35 can influence vascular inflammation and atherosclerosis has however never been tested. Lethally irradiated LdlrKO mice were randomized to receive GPR35KO or wild type (WT) bone marrow transplants and fed a high cholesterol diet for eight weeks to develop atherosclerosis. GPR35KO and WT chimeric mice presented no difference in the size of atherosclerotic lesions in the aortic arch (2.37 ± 0.58% vs. 1.95 ± 0.46%, respectively) or in the aortic roots (14.77 ± 3.33% vs. 11.57 ± 2.49%, respectively). In line with these data, no changes in the percentage of VCAM-1+, IAb + cells, and CD3+ T cells, as well as alpha smooth muscle cell actin expression, was observed between groups. Interestingly, the GPR35KO group presented a small but significant increase in CD68+ macrophage infiltration in the plaque. However, in vitro culture experiments using bone marrow-derived macrophages from both groups indicated that GPR35 plays no role in modulating the secretion of major inflammatory cytokines. Our study indicates that GPR35 expression does not play a direct role in macrophage activation, vascular inflammation, and the development of atherosclerosis.
- Published
- 2021
- Full Text
- View/download PDF
39. N -acyl taurines are endogenous lipid messengers that improve glucose homeostasis
- Author
-
Filip K. Knop, Matthew P. Gillum, Samuel A.J. Trammell, Richard G. Kibbey, Christina Nexøe-Larsen, Jens S. Svenningsen, Benjamin F. Cravatt, Michele K. McKinney, Daisuke Ogasawara, Natalia Petersen, Rebecca L. Cardone, Trisha J. Grevengoed, and Thue W. Schwartz
- Subjects
chemistry.chemical_classification ,endocrine system ,Taurine ,Multidisciplinary ,fungi ,Glucagon secretion ,Fatty acid ,Lipid signaling ,Metabolism ,Biological Sciences ,body regions ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Fatty acid amide hydrolase ,Glucose homeostasis ,lipids (amino acids, peptides, and proteins) ,Blood sugar regulation - Abstract
Fatty acid amide hydrolase (FAAH) degrades 2 major classes of bioactive fatty acid amides, the N-acylethanolamines (NAEs) and N-acyl taurines (NATs), in central and peripheral tissues. A functional polymorphism in the human FAAH gene is linked to obesity and mice lacking FAAH show altered metabolic states, but whether these phenotypes are caused by elevations in NAEs or NATs is unknown. To overcome the problem of concurrent elevation of NAEs and NATs caused by genetic or pharmacological disruption of FAAH in vivo, we developed an engineered mouse model harboring a single-amino acid substitution in FAAH (S268D) that selectively disrupts NAT, but not NAE, hydrolytic activity. The FAAH-S268D mice accordingly show substantial elevations in NATs without alterations in NAE content, a unique metabolic profile that correlates with heightened insulin sensitivity and GLP-1 secretion. We also show that N-oleoyl taurine (C18:1 NAT), the most abundant NAT in human plasma, decreases food intake, improves glucose tolerance, and stimulates GPR119-dependent GLP-1 and glucagon secretion in mice. Together, these data suggest that NATs act as a class of lipid messengers that improve postprandial glucose regulation and may have potential as investigational metabolites to modify metabolic disease.
- Published
- 2019
40. The Molecular Diversity of Vagal Afferents Revealed
- Author
-
Thue W. Schwartz, Kristoffer L. Egerod, and Laurent Gautron
- Subjects
0301 basic medicine ,General Neuroscience ,Cell ,RNA ,Biology ,Vagus nerve ,Transcriptome ,03 medical and health sciences ,Autonomic nervous system ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Using single-cell RNA sequencing (RNA-seq), Kupari and coworkers (Cell Rep., 2019) have generated a long sought-after molecular atlas of vagal afferents in the mouse. Vagal afferents were found to be organized into 24 subtypes, revealing a level of diversity that was not previously recognized.
- Published
- 2019
41. Microbial fermentation of flaxseed fibers modulates the transcriptome of GPR41-expressing enteroendocrine cells and protects mice against diet-induced obesity
- Author
-
Mette Kristensen, Rozita Akrami, Petia Kovatcheva-Datchary, Tulika Arora, Thue W. Schwartz, Fredrik Bäckhed, Olga Rudenko, Anna Sofie Husted, and Kristoffer L. Egerod
- Subjects
Dietary Fiber ,Male ,0301 basic medicine ,Physiology ,Endocrinology, Diabetes and Metabolism ,Enteroendocrine cell ,Gut flora ,Receptors, G-Protein-Coupled ,Transcriptome ,Mice ,0302 clinical medicine ,Flax ,Cecum ,2. Zero hunger ,chemistry.chemical_classification ,biology ,digestive, oral, and skin physiology ,Biochemistry ,Female ,medicine.medical_specialty ,Colon ,Enteroendocrine Cells ,Firmicutes ,030209 endocrinology & metabolism ,Diet, High-Fat ,digestive system ,03 medical and health sciences ,Verrucomicrobia ,Ileum ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Obesity ,Microbiome ,Cellulose ,Metabolism ,Fatty Acids, Volatile ,biology.organism_classification ,medicine.disease ,Gastrointestinal Microbiome ,Lactobacillus ,030104 developmental biology ,Enzyme ,Endocrinology ,chemistry ,Fermentation ,Bifidobacterium - Abstract
Dietary fibers, an integral part of the human diet, require the enzymatic activity of the gut microbiota for complete metabolism into short-chain fatty acids (SCFAs). SCFAs are important modulators of host metabolism and physiology and act in part as signaling molecules by activating G protein-coupled receptors (GPCRs), such as GPR41. Flaxseed fibers improve metabolism in rodents and mice, but their fermentation profiles, effects on enteroendocrine cells, and associated metabolic benefits are unknown. We fed GPR41-red fluorescent protein mice, an enteroendocrine reporter mouse strain, chow, high-fat diet (HFD), or HFD supplemented either with 10% nonfermentable fiber cellulose or fermentable flaxseed fibers for 12 wk to assess changes in cecal gut microbiota, enteroendocrine cell transcriptome in the ileum and colon, and physiological parameters. We observed that flaxseed fibers restructured the gut microbiota and promoted proliferation of the genera Bifidobacterium and Akkermansia compared with HFD. The shifts in cecal bacterial composition restored levels of the SCFAs butyrate similar to the chow diet, resulting in colonic but not ileal enteroendocrine cell transcriptional changes in genes related to cell cycle, mRNA, and protein transport compared with HFD. Consistent with the effects on enteroendocrine functions, flaxseed fibers also protected mice from diet-induced obesity, potentially by preventing a reduction in energy expenditure induced by an HFD. Our study shows that flaxseed fibers alter cecal microbial ecology, are fermented to SCFAs in the cecum, and modulate enteroendocrine cell transcriptome in the colon, which may contribute to their metabolically favorable phenotype.
- Published
- 2019
42. Preassociation between the 5‐HT 7 serotonin receptor and G protein G s : molecular determinants and association with low potency activation of adenylyl cyclase
- Author
-
Finn Olav Levy, Kjetil Wessel Andressen, Andrea Hembre Ulsund, Marie Dahl, Thue W. Schwartz, Thomas M. Frimurer, and Ornella Manfra
- Subjects
0301 basic medicine ,Agonist ,medicine.drug_class ,G protein ,Fluorescence recovery after photobleaching ,Biochemistry ,Cell biology ,Adenylyl cyclase ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Genetics ,medicine ,Receptor ,Molecular Biology ,030217 neurology & neurosurgery ,5-HT receptor ,Intracellular ,Biotechnology ,G protein-coupled receptor - Abstract
According to early models of GPCR signaling, G proteins only interact with activated receptors. However, some GPCRs were shown to assemble with G proteins before receptor activation, in accordance with more recent models. Previously, we found that the 5-HT7 receptor, as opposed to the 5-HT4 receptor, was preassociated with Gs, but the molecular determinants for this interaction are still elusive. In a series of chimeric 5-HT7 receptors with intracellular segments from 5-HT4, we determined the receptor-G protein interaction by performing antibody-immobilized fluorescence recovery after photobleaching and fluorescence resonance energy transfer. We identified the intracellular loop 3 and C-tail of the 5-HT7 receptor to be responsible for the preassociation with Gs, and we further delineated the TM5 extension in the intracellular loop 3 and helix 8 in the C-tail as the molecular determinants. These chimeric exchanges converted the 5-HT7 receptor into a collision-coupled receptor that recruited G proteins only upon agonist activation, whereas reciprocal exchanges converted 5-HT4 to a preassociated receptor. The 5-HT7 receptor displayed 2-component agonist-induced Gs signaling with high and low potency. In addition, the same segments were involved in low-potency signaling and preassociation. The correspondence between Gs preassociation and low-potency Gs signaling is a novel aspect of GPCR pharmacology.-Ulsund, A. H., Dahl, M., Frimurer, T. M., Manfra, O., Schwartz, T. W., Levy, F. O., Andressen, K. W. Preassociation between the 5-HT7 serotonin receptor and G protein Gs: molecular determinants and association with low potency activation of adenylyl cyclase.
- Published
- 2018
43. A systems biology approach to understand gut microbiota and host metabolism in morbid obesity: design of the BARIA Longitudinal Cohort Study
- Author
-
N. C. Bosma, Ömrüm Aydin, Louise E. Olofsson, Fredrik Bäckhed, Siv A. Hjorth, N. van Olst, C. C. Van Olden, J. Verheij, Hilde Herrema, M. Nieuwdorp, Jens Nielsen, A. W. van de Laar, Sjoerd C. Bruin, V. E. A. Gerdes, Dimitra Lappa, Mariët Hagedoorn, Abraham S. Meijnikman, Thue W. Schwartz, Albert K. Groen, Annika Lundqvist, L. M. de Brauw, Valentina Tremaroli, Yair I. Z. Acherman, Robbert Sanderman, J. B. Hoozemans, Johanna Eveliina Pyykkö, Graduate School, ACS - Atherosclerosis & ischemic syndromes, ACS - Diabetes & metabolism, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, Paediatric Surgery, Vascular Medicine, Pathology, and Experimental Vascular Medicine
- Subjects
0301 basic medicine ,Male ,obesity ,SURGERY ,medicine.medical_treatment ,Physiology ,Y GASTRIC BYPASS ,030204 cardiovascular system & hematology ,0302 clinical medicine ,Weight loss ,insulin resistance ,Glucose homeostasis ,Insulin ,Longitudinal Studies ,metabolites ,GENE-EXPRESSION ,Netherlands ,OUTCOMES ,INSULIN SENSITIVITY ,Systems Biology ,Fatty liver ,Middle Aged ,Obesity, Morbid ,Postprandial ,Phenotype ,Research Design ,ADIPOSITY ,Original Article ,Female ,medicine.symptom ,Adult ,bariatric surgery ,03 medical and health sciences ,Insulin resistance ,Internal Medicine ,medicine ,Humans ,Triglycerides ,RESTORATION ,gut microbiota ,business.industry ,Type 2 Diabetes Mellitus ,Original Articles ,DRIVEN ,medicine.disease ,Obesity ,Gastrointestinal Microbiome ,Fatty Liver ,030104 developmental biology ,Glucose ,WEIGHT MAINTENANCE ,GLUCOSE-TOLERANCE ,business ,Biomarkers - Abstract
Introduction Prevalence of obesity and associated diseases, including type 2 diabetes mellitus, dyslipidaemia and non‐alcoholic fatty liver disease (NAFLD), are increasing. Underlying mechanisms, especially in humans, are unclear. Bariatric surgery provides the unique opportunity to obtain biopsies and portal vein blood‐samples. Methods The BARIA Study aims to assess how microbiota and their metabolites affect transcription in key tissues and clinical outcome in obese subjects and how baseline anthropometric and metabolic characteristics determine weight loss and glucose homeostasis after bariatric surgery. We phenotype patients undergoing bariatric surgery (predominantly laparoscopic Roux‐en‐Y gastric bypass), before weight loss, with biometrics, dietary and psychological questionnaires, mixed meal test (MMT) and collect fecal‐samples and intra‐operative biopsies from liver, adipose tissues and jejunum. We aim to include 1500 patients. A subset (approximately 25%) will undergo intra‐operative portal vein blood‐sampling. Fecal‐samples are analyzed with shotgun metagenomics and targeted metabolomics, fasted and postprandial plasma‐samples are subjected to metabolomics, and RNA is extracted from the tissues for RNAseq‐analyses. Data will be integrated using state‐of‐the‐art neuronal networks and metabolic modeling. Patient follow‐up will be ten years. Results Preoperative MMT of 170 patients were analysed and clear differences were observed in glucose homeostasis between individuals. Repeated MMT in 10 patients showed satisfactory intra‐individual reproducibility, with differences in plasma glucose, insulin and triglycerides within 20% of the mean difference. Conclusion The BARIA study can add more understanding in how gut‐microbiota affect metabolism, especially with regard to obesity, glucose metabolism and NAFLD. Identification of key factors may provide diagnostic and therapeutic leads to control the obesity‐associated disease epidemic.
- Published
- 2021
44. Extracellular succinate hyperpolarizes M2 macrophages through SUCNR1/GPR91-mediated Gq signaling
- Author
-
Thomas K. Hiron, Siv A. Hjorth, Anna Sofie Husted, Michel Bouvier, Da Lin, Jacob E. Petersen, Billy Breton, Mette Trauelsen, Christopher A. O’Callaghan, Asuka Inoue, Thue W. Schwartz, and Thomas M. Frimurer
- Subjects
0301 basic medicine ,Male ,Transcriptional Activation ,G protein ,Arrestins ,QH301-705.5 ,GPR91 ,Succinic Acid ,GTP-Binding Protein alpha Subunits, Gi-Go ,Ligands ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Transcriptional regulation ,Extracellular ,Humans ,M2 macrophages ,Biology (General) ,Receptor ,Gq signaling ,Chemistry ,Macrophages ,non-metabolite ligands ,succinate ,Phenotype ,Cell biology ,Citric acid cycle ,Protein Subunits ,030104 developmental biology ,Gene Ontology ,HEK293 Cells ,Gene Expression Regulation ,SUCNR1 ,Type C Phospholipases ,GTP-Binding Protein alpha Subunits, Gq-G11 ,Female ,Signal transduction ,Extracellular Space ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Summary: Succinate functions both as a classical TCA cycle metabolite and an extracellular metabolic stress signal sensed by the mainly Gi-coupled succinate receptor SUCNR1. In the present study, we characterize and compare effects and signaling pathways activated by succinate and both classes of non-metabolite SUCNR1 agonists. By use of specific receptor and pathway inhibitors, rescue in G-protein-depleted cells and monitoring of receptor G protein activation by BRET, we identify Gq rather than Gi signaling to be responsible for SUCNR1-mediated effects on basic transcriptional regulation. Importantly, in primary human M2 macrophages, in which SUCNR1 is highly expressed, we demonstrate that physiological concentrations of extracellular succinate act through SUCNR1-activated Gq signaling to efficiently regulate transcription of immune function genes in a manner that hyperpolarizes their M2 versus M1 phenotype. Thus, sensing of stress-induced extracellular succinate by SUCNR1 is an important transcriptional regulator in human M2 macrophages through Gq signaling.
- Published
- 2021
45. Autocrine negative feedback regulation of lipolysis through sensing of NEFAs by FFAR4/GPR120 in WAT
- Author
-
Max Nieuwdorp, Lars O. Dragsted, Trond Ulven, Stijn Meijnikman, Jeppe H. Ekberg, Sjoerd C. Bruin, Zach Gerhart-Hines, Yair I. Z. Acherman, Shannon L. O'Brien, Thue W. Schwartz, Tinne A.D. Nissen, Davide Calebiro, Emma Tripp, Anna Sofie Husted, Experimental Vascular Medicine, Vascular Medicine, ACS - Diabetes & metabolism, and AGEM - Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
0301 basic medicine ,Male ,GPR120 ,Adipocytes, White ,Fatty Acids, Nonesterified ,NEFA ,Mass Spectrometry ,Receptors, G-Protein-Coupled ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,GPCR ,Adipocyte ,Adipocytes ,Receptor ,Feedback, Physiological ,Fatty Acids ,Autocrine Communication ,Nicotinic agonist ,Adipose Tissue ,Female ,Agonist ,medicine.medical_specialty ,lcsh:Internal medicine ,medicine.drug_class ,G protein ,Adipose Tissue, White ,Lipolysis ,030209 endocrinology & metabolism ,Brief Communication ,03 medical and health sciences ,FFAR4 ,Internal medicine ,3T3-L1 Cells ,medicine ,Animals ,Humans ,Autocrine ,Autocrine signalling ,lcsh:RC31-1245 ,Molecular Biology ,Cell Biology ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,chemistry ,Culture Media, Conditioned ,Chromatography, Liquid - Abstract
Objectives Long-chain fatty acids (LCFAs) released from adipocytes inhibit lipolysis through an unclear mechanism. We hypothesized that the LCFA receptor, FFAR4 (GPR120), which is highly expressed in adipocytes, may be involved in this feedback regulation. Methods and results Liquid chromatography mass spectrometry (LC-MS) analysis of conditioned media from isoproterenol-stimulated primary cultures of murine and human adipocytes demonstrated that most of the released non-esterified free fatty acids (NEFAs) are known agonists for FFAR4. In agreement with this, conditioned medium from isoproterenol-treated adipocytes stimulated signaling strongly in FFAR4 transfected COS-7 cells as opposed to non-transfected control cells. In transfected 3T3-L1 cells, FFAR4 agonism stimulated Gi- and Go-mini G protein binding more strongly than Gq, effects which were blocked by the selective FFAR4 antagonist AH7614. In primary cultures of murine white adipocytes, the synthetic, selective FFAR4 agonist CpdA inhibited isoproterenol-induced intracellular cAMP accumulation in a manner similar to the antilipolytic control agent nicotinic acid acting through another receptor, HCAR2. In vivo, oral gavage with the synthetic, specific FFAR4 agonist CpdB decreased the level of circulating NEFAs in fasting lean mice to a similar degree as nicotinic acid. In agreement with the identified anti-lipolytic effect of FFAR4, plasma NEFAs and glycerol were increased in FFAR4-deficient mice as compared to littermate controls despite having elevated insulin levels, and cAMP accumulation in primary adipocyte cultures was augmented by treatment with the FFAR4 antagonist conceivably by blocking the stimulatory tone of endogenous NEFAs on FFAR4. Conclusions In white adipocytes, FFAR4 functions as an NEFA-activated, autocrine, negative feedback regulator of lipolysis by decreasing cAMP though Gi-mediated signaling., Graphical abstract Image 1Simplified schematic overview of control of lipolysis by GPCRs with focus on FFAR4 in the context of the other putative autocrine feedback mechanisms. The left side indicates the autocrine NEFA and FFAR4 and Gi-mediated autocrine negative feedback loop on lipolysis characterized in the present study, including the pharmacological FFAR4 tool compounds used. The classical catecholamine, β1-receptor, Gs, and adenylate cyclase (AC)-mediated stimulatory control of lipolysis are indicated in green. The right side indicates six different Gi-coupled metabolite sensing GPCRs, which all act as autocrine regulators of lipolysis and other adipocyte functions under different physiological circumstances: The adenosine A1 receptor, which is responsible for the original, classical autocrine effects of adenosine [52]; the lactate receptor, HCAR1/GPR81, which is responsible for the insulin, glucose-mediated inhibition of lipolysis [47]; the HCAR2/GPR109A receptor for circulating and possibly also locally produced β-hydroxybutyrate and the prototype anti-lipolytic drug niacin [53]; the HCAR3/GPR109B receptor for β-hydroxyoctanoate [54]; the SUCNR1/GPR91 receptor sensing stress and hypoxia-induced succinate excretion [39]; and FFAR2/GPR43, which senses acetate conceivably both through autocrine and paracrine mechanisms from certain subtypes of adipocytes [55]., Highlights • Conditioned medium from adipocytes contains multiple FFAR4 agonist NEFAs. • Conditioned medium from adipocytes stimulates FFAR4 signaling. • Synthetic FFAR4 agonist stimulates Gi in vitro and inhibits lipolysis in vivo. • Synthetic FFAR4 antagonist increases adipocyte cAMP by blocking NEFA tone. • Circulating NEFAs and glycerol are elevated in FFAR4 KO animals despite high insulin levels.
- Published
- 2020
46. Post-oral fat-induced satiation is mediated by endogenous CCK and GLP-1 in a fat self-administration mouse model
- Author
-
Jeppe H. Ekberg, Birgit L. Lindberg, Thue W. Schwartz, Karen Kleberg, Vasiliki Vana, Piotr A. Mroz, Michelle K. Lærke, Harald S. Hansen, and Jens F. Rehfeld
- Subjects
medicine.medical_specialty ,medicine.drug_class ,Dietary lipid ,Experimental and Cognitive Psychology ,Devazepide ,Satiation ,03 medical and health sciences ,Behavioral Neuroscience ,Eating ,Mice ,0302 clinical medicine ,Glucagon-Like Peptide 1 ,Internal medicine ,medicine ,Animals ,0501 psychology and cognitive sciences ,Peptide YY ,050102 behavioral science & comparative psychology ,Receptor ,Glucagon-like peptide 1 receptor ,Cholecystokinin ,Chemistry ,digestive, oral, and skin physiology ,05 social sciences ,Antagonist ,Receptor antagonist ,Endocrinology ,Receptors, Cholecystokinin ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery - Abstract
Triacylglycerol is the most abundant dietary lipid, and a strong stimulator of satiation. Absorption of triacylglycerol in the small intestine occurs in the form of free fatty acids and 2-monoacylglycerol, a process known to trigger not only the release of cholecystokinin (CCK) but also glucagon-like peptide 1 (GLP-1) and peptide YY (PYY). It remains controversial, however, whether endogenously released GLP-1 and PYY are required for fat-induced satiation. Using a self-administration model where mice are trained to self-administer Intralipid 30% intragastrically, we show that blocking the CCK1 receptors with intraperitoneal devazepide diminishes the post-oral satiation effect of ingested fat. Similarly, s.c. administration of a GLP-1 receptor antagonist with a prolonged half-life (Jant4-C16) also reduced the post-oral satiation effect of ingested fat. Importantly, coadministration of the GLP-1 antagonist together with devazepide increased fat self-infusions to a level equal to the combined blockade of each individual peptide action alone, indicating an additive effect of endogenous CCK and GLP-1 in fat satiation signaling. Blocking the PYY Y2 receptor did not further enhance the fat intake in devazepide-treated mice. Consistent with the above, we show that voluntary post-oral ingestion of fat increases CCK and GLP-1 plasma levels and is correlated positively with CCK and GLP-1 plasma concentrations. Taken together, our results support the role of endogenous GLP-1 in the regulation of fat intake and suggest that both CCK and GLP-1 are required for the fat satiation signaling.
- Published
- 2020
47. Synthetic G protein‐coupled bile acid receptor agonists and bile acids act via basolateral receptors in ileal and colonic mucosa
- Author
-
Iain R. Tough, Helen M. Cox, and Thue W. Schwartz
- Subjects
Male ,0301 basic medicine ,Agonist ,Mice, 129 Strain ,Colon ,Physiology ,medicine.drug_class ,030209 endocrinology & metabolism ,fecal pellet transit ,Receptors, G-Protein-Coupled ,Receptors, Gastrointestinal Hormone ,Nitric oxide ,Bile Acids and Salts ,G protein-coupled bile acid receptor ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Organ Culture Techniques ,0302 clinical medicine ,Ileum ,peptide YY ,medicine ,Animals ,Humans ,Intestinal Mucosa ,Receptor ,Aged ,Mice, Knockout ,Lamina propria ,Bile acid ,Endocrine and Autonomic Systems ,Chemistry ,digestive, oral, and skin physiology ,Gastroenterology ,Middle Aged ,Molecular biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Peptide YY ,Enterochromaffin cell ,Female ,colonic ion transport - Abstract
Background The G protein-coupled bile acid (BA) receptor, GPBA (previously named TGR5), mediates BA gastrointestinal (GI) activities. Our aim was to elucidate the mucosal and motility responses to selective GPBA agonists compared with conjugated BA (eg, taurodeoxycholate, TDCA) in mouse and human colon. Methods Ion transport responses to GPBA agonists or BAs were measured in mucosal preparations with intact submucous innervation, from C57Bl/6, PYY-/-, or GPBA-/- mice and compared with GPBA signaling in human colon. We also investigated the mechanisms underlying GPBA agonism in mucosae and on natural fecal pellet propulsion. Key results GPBA agonist Merck V stimulated basolateral responses involving peptide YY (PYY), cholinergic, and 5-HT mechanisms in colonic mucosa. The PYY-mediated GPBA signal was glucose-sensitive. Luminal TDCA crossed the epithelial lining via the apical sodium-dependent BA transporter (ASBT) and its inhibitor, GSK2330672 significantly reduced luminal, but not basolateral TDCA activity. Merck V also slowed natural fecal pellet progression in wild-type and PYY-/- colons but not in GPBA-/- colon, while TDCA increased motility in wild-type colon. The antimotile GPBA effect was reversed by blockade of glucagon-like peptide 1 (GLP-1) receptors or nitric oxide synthase, indicating involvement of GLP-1 and nitric oxide. Conclusions & inferences We conclude that several different targets within the lamina propria express GPBA, including L cells (that release PYY and GLP-1), enterochromaffin cells and neurons (that release 5-HT), and other enteric neurons. Furthermore, luminal-conjugated BAs require transport across the epithelium via ASBT in order to activate basolateral GPBA.
- Published
- 2020
48. Why Warburg Works: Lactate Controls Immune Evasion through GPR81
- Author
-
Thue W. Schwartz, Mette Trauelsen, Kathrine Lundø, and Stine F. Pedersen
- Subjects
0301 basic medicine ,Stromal cell ,Physiology ,Antigen-Presenting Cells ,Breast Neoplasms ,GPR81 ,Biology ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,Humans ,Lactic Acid ,Autocrine signalling ,Molecular Biology ,Immune Evasion ,Cell Biology ,Metabolism ,Evasion (ethics) ,3. Good health ,030104 developmental biology ,Cancer cell ,Cancer research ,030217 neurology & neurosurgery - Abstract
Lactate accumulation in tumors—a hallmark of the Warburg effect—has recently been shown to regulate cancer cell metabolism and survival through autocrine activation of GPR81. Now, Brown et al. (2020) demonstrate that lactate surprisingly also controls immune evasion through paracrine activation of GPR81 on stromal dendritic cells.
- Published
- 2020
49. Structural basis for GPCR signaling by small polar versus large lipid metabolites-discovery of non-metabolite ligands
- Author
-
Michael Lückmann, Mette Trauelsen, Thue W. Schwartz, and Thomas M. Frimurer
- Subjects
0303 health sciences ,Metabolite ,Cell ,Transporter ,Cell Biology ,Biology ,Ligands ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Biochemistry ,Free fatty acid receptor 1 ,medicine ,Extracellular ,Humans ,Lipid bilayer ,Receptor ,030217 neurology & neurosurgery ,030304 developmental biology ,G protein-coupled receptor ,Signal Transduction - Abstract
Key metabolites act through specific G protein-coupled receptors (GPCRs) as extracellular signals of fuel availability and metabolic stress. Here, we focus on the succinate receptor SUCNR1/GPR91 and the long chain fatty acid receptor FFAR1/GPR40, for which 3D structural information is available. Like other small polar acidic metabolites, succinate is excreted from the cell by transporter proteins to bind to an extracellular, solvent-exposed pocket in SUCNR1. Non-metabolite pharmacological tool compounds are currently being designed based on the structure of the SUCNR1 binding pocket. In FFAR1, differently signaling lipid mimetics bind in two distinct membrane-exposed sites corresponding to each of the lipid bilayer leaflets. Conceivably endogenous lipid ligands gain access to these sites by way of the membrane and probably occupy both sites under physiological circumstances. Design of polar agonists for a dynamic, solvent-exposed pocket in FFAR1 underlines the possibility of structure-based approaches for development of novel tool compounds even in lipid sensing metabolite GPCRs.
- Published
- 2019
50. Structure-Activity Investigations and Optimisations of Non-metabolite Agonists for the Succinate Receptor 1
- Author
-
Line Ørsted Bielefeldt, Mette Trauelsen, Elisabeth Rexen Ulven, Matjaz Brvar, Lisa K. I. Jensen, Michael Lückmann, Thue W. Schwartz, and Thomas M. Frimurer
- Subjects
0301 basic medicine ,Agonist ,medicine.drug_class ,Metabolite ,Succinic Acid ,Adipose tissue ,lcsh:Medicine ,Crystallography, X-Ray ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Receptors, Purinergic P2Y1 ,Structure-Activity Relationship ,medicine ,Succinate receptor 1 ,Potency ,Animals ,Humans ,Receptor ,lcsh:Science ,Kidney ,Multidisciplinary ,lcsh:R ,In vitro toxicology ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry ,chemistry ,lcsh:Q - Abstract
The succinate receptor 1 (SUCNR1) is a receptor for the metabolite succinate, which functions as a metabolic stress signal in the liver, kidney, adipose tissue and the retina. However, potent non-metabolite tool compounds are needed to reveal the physiological role and pharmacological potential of SUCNR1. Recently, we published the discovery of a computationally receptor-structure derived non-metabolite SUCNR1 agonist series with high target selectivity. We here report our structure-activity exploration and optimisation that has resulted in the development of agonists with nanomolar potency and excellent solubility and stability properties in a number of in vitro assays. Ligand-guided receptor models with high discriminative power between binding of active and inactive compounds were developed for design of novel chemotypes.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.