215 results on '"Pedersen SF"'
Search Results
2. Development of complex model systems for analysis of cell-cell and cell-microenvironment interactions in breast cancer
- Author
-
Andersen, AP, Ronnov-Jessen, L, Hulikova, A, Swietach, P, and Pedersen, SF
- Published
- 2016
3. HIV/SIV escape from immune surveillance: focus on Nef
- Author
-
Tolstrup M, Ostergaard L, Laursen AL, Pedersen SF, Duch M
- Published
- 2004
4. Microvessel density but not neoangiogenesis is associated with 18F-FDG uptake in human atherosclerotic carotid plaques.
- Author
-
Pedersen SF, Graebe M, Hag AM, Hoejgaard L, Sillesen H, Kjaer A, Pedersen, Sune Folke, Graebe, Martin, Hag, Anne Mette Fisker, Hoejgaard, Liselotte, Sillesen, Henrik, and Kjaer, Andreas
- Abstract
Introduction: The vulnerable atherosclerotic lesion exhibits the proliferation of neovessels and inflammation. The imaging modality 2-deoxy-2-[(18)F]fluoro-D: -glucose positron emission tomography ((18)FDG-PET) is considered for the identification of vulnerable plaques.Purpose: The purpose of this study was to compare the gene expression of neoangiogenesis and vulnerability-associated genes with (18)FDG uptake in patients undergoing carotid endarterectomy.Procedures: Human atherosclerotic carotid artery plaques from symptomatic patients were used for gene expression analysis by quantitative PCR of vascular endothelial growth factor (VEGF) and integrin α(V) and integrin β(3) subunits, genes essential during neoangiogenesis. We also evaluated the gene expression of CD34, a measure of microvessel density (MVD), as well as CD68, MMP-9, and cathepsin K, genes of major importance in plaque vulnerability. Gene expression analysis was compared with (18)FDG-PET.Results: VEGF and integrin α(V)β(3) gene expression did not correlate with (18)FDG uptake, whereas CD34 gene expression exhibited an inverse correlation with (18)FDG uptake. Additionally, we established that markers of vulnerability were correlated with (18)FDG uptake.Conclusions: Neoangiogenesis is not associated with (18)FDG uptake, whereas MVD and markers of vulnerability correlate with (18)FDG uptake. The absence of correlation between markers of neoangiogenesis and (18)FDG uptake suggests a temporal separation between the process of neoangiogenesis and inflammatory activity. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
5. Determination of edema in porcine coronary arteries by T2 weighted cardiovascular magnetic resonance.
- Author
-
Pedersen SF, Thrysøe SA, Paaske WP, Thim T, Falk E, Ringgaard S, and Kim WY
- Abstract
BACKGROUND:Inflammation plays a pivotal role in all stages of atherosclerosis. Since edema is known to be an integral part of inflammation, a noninvasive technique that can identify edema in the coronary artery wall may provide unique information regarding plaque activity. In this study, we aimed to determine whether edema induced in porcine coronary arteries by balloon injury could be reliably detected by cardiovascular magnetic resonance (CMR) using a water sensitive T2-weighted short tau inversion recovery sequence (T2-STIR). We also aimed to compare these results to those of conventional T2-weighted (T2W) imaging.METHODS:Edema was induced in the proximal left anterior descending (LAD) coronary artery wall in seven pigs by balloon injury. At baseline, and 1-10 days (average four) post injury, the proximal LAD was assessed by water sensitive T2-STIR and conventional T2W sequences in cross-sectional planes. CMR images were matched to histopathology, validated against Evans blue as a marker of increased vessel wall permeability, and correlated with the arterial amount of fibrinogen used as an edema surrogate marker.RESULTS:Post injury, the T2-STIR images of the injured LAD vessel wall showed a significant 72%, relative signal intensity (SI) increase compared with baseline (p = 0.028). Using a threshold value of SI 7 SD above the average SI of the myocardium, T2-STIR detected edema in the vessel wall (i.e. enhancement) with a sensitivity of 100 and a specificity of 71. Twelve out of the 14 (86%) T2-STIR images displaying coronary artery wall enhancement also showed Evans blue uptake in the corresponding histology. The relative signal intensity showed a linear correlation with the amount of fibrinogen detected on the corresponding histopathology (? = 0.750, p = 0.05). The conventional T2W images did not show significant changes in SI post injury.CONCLUSION:T2-STIR CMR enabled detection of coronary artery wall edema and could therefore be a non-invasive diagnostic tool for evaluation of inflammatory coronary artery wall activity. [ABSTRACT FROM AUTHOR]
- Published
- 2011
6. The Na+/H+ exchanger NHE1, but not the Na+, HCO3(-) cotransporter NBCn1, regulates motility of MCF7 breast cancer cells expressing constitutively active ErbB2.
- Author
-
Lauritzen G, Stock CM, Lemaire J, Lund SF, Jensen MF, Damsgaard B, Petersen KS, Wiwel M, Rønnov-Jessen L, Schwab A, Pedersen SF, Lauritzen, Gitte, Stock, Christian-Martin, Lemaire, Justine, Lund, Stine F, Jensen, Mie Frid, Damsgaard, Britt, Petersen, Katrine Seide, Wiwel, Maria, and Rønnov-Jessen, Lone
- Abstract
We and others have shown central roles of the Na(+)/H(+) exchanger NHE1 in cell motility. The aim of this study was to determine the roles of NHE1 and of the Na(+), HCO(3)(-) cotransporter NBCn1 in motility of serum-starved MCF-7 breast cancer cells expressing constitutively active ErbB2 (ΔNErbB2). ΔNErbB2 expression elicited NBCn1 upregulation, Ser(703)-phosphorylation of NHE1, and NHE1-inhibitor (EIPA)-sensitive pericellular acidification, in conjunction with increased expression of β1 integrin and ERM proteins. Active ERM proteins and NHE1 colocalized strongly to invadopodial rosettes, the diameter of which was increased by ΔNErbB2. Adhesion and migration on collagen-I were augmented by ΔNErbB2, unaffected by the NBC inhibitor S0859, and further stimulated by EIPA in a manner potentiated by PI3K-Akt-inhibition. These findings demonstrate that NHE1 inhibition can enhance cancer cell motility, adding an important facet to the understanding of NHE1 in cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
7. Mimicking and analyzing the tumor microenvironment.
- Author
-
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, and Pedersen SF
- Subjects
- Humans, Animals, Models, Biological, Tumor Microenvironment, Neoplasms pathology, Neoplasms genetics, Neoplasms therapy
- Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges., Competing Interests: Declaration of interests S.F.P. is a cofounder of SOLID Therapeutics., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. NHE1 regulation in NAFLD in vitro contributes to hepatocyte injury and HSC crosstalk.
- Author
-
Sjøgaard-Frich LM, Henriksen MS, Lam SM, Birkbak FJ, Czaplinska D, Flinck M, and Pedersen SF
- Subjects
- Humans, Animals, Mice, Hep G2 Cells, Sodium-Hydrogen Exchanger 1 metabolism, Sodium-Hydrogen Exchanger 1 genetics, Hepatocytes metabolism, Non-alcoholic Fatty Liver Disease metabolism, Non-alcoholic Fatty Liver Disease genetics, Hepatic Stellate Cells metabolism
- Abstract
Non-alcoholic fatty liver disease (NAFLD) is the fastest-growing cause of liver-associated death globally. Whole-body knockout (KO) of Na+/H+ exchanger 1 (NHE1, SLC9A1) was previously proposed to protect against high-fat diet-induced liver damage; however, mechanistic insight was lacking. The aim of the present work was to address this question in vitro to determine how NHE1, specifically in hepatocytes, impacts lipid overload-induced inflammation, fibrosis, and hepatocyte-hepatic stellate cell (HSC) crosstalk. We induced palmitate (PA)-based steatosis in AML12 and HepG2 hepatocytes; manipulated NHE1 activity pharmacologically and by CRISPR/Cas9-mediated KO and overexpression; and measured intracellular pH (pHi), steatosis-associated inflammatory and fibrotic mediators, and cell death. PA treatment increased NHE1 mRNA levels but modestly reduced NHE1 protein expression and hepatocyte pHi. NHE1 KO in hepatocytes did not alter lipid droplet accumulation but reduced inflammatory signaling (p38 MAPK activity), lipotoxicity (4-HNE accumulation), and apoptosis (poly-ADP-ribose-polymerase-1 (PARP) cleavage). Conditioned medium from PA-treated hepatocytes increased the expression of NHE1 and of the fibrosis regulator tissue inhibitor of matrix metalloproteinases-2 in LX-2 HSCs, in a manner abolished by NHE1 KO in hepatocytes. We conclude that NHE1 is regulated in NAFLD in vitro and contributes to the ensuing damage by aggravating hepatocyte injury and stimulating hepatocyte-HSC crosstalk.
- Published
- 2024
- Full Text
- View/download PDF
9. Regulation of cancer cell lipid metabolism and oxidative phosphorylation by microenvironmental acidosis.
- Author
-
Rolver MG, Severin M, and Pedersen SF
- Subjects
- Humans, Animals, Mitochondria metabolism, Mitochondria pathology, Signal Transduction, Oxidative Stress, Oxidative Phosphorylation, Tumor Microenvironment, Acidosis metabolism, Acidosis pathology, Lipid Metabolism physiology, Neoplasms metabolism, Neoplasms pathology
- Abstract
The expansion of cancer cell mass in solid tumors generates a harsh environment characterized by dynamically varying levels of acidosis, hypoxia, and nutrient deprivation. Because acidosis inhibits glycolytic metabolism and hypoxia inhibits oxidative phosphorylation, cancer cells that survive and grow in these environments must rewire their metabolism and develop a high degree of metabolic plasticity to meet their energetic and biosynthetic demands. Cancer cells frequently upregulate pathways enabling the uptake and utilization of lipids and other nutrients derived from dead or recruited stromal cells, and in particular lipid uptake is strongly enhanced in acidic microenvironments. The resulting lipid accumulation and increased reliance on β-oxidation and mitochondrial metabolism increase susceptibility to oxidative stress, lipotoxicity, and ferroptosis, in turn driving changes that may mitigate such risks. The spatially and temporally heterogeneous tumor microenvironment thus selects for invasive, metabolically flexible, and resilient cancer cells capable of exploiting their local conditions and of seeking out more favorable surroundings. This phenotype relies on the interplay between metabolism, acidosis, and oncogenic mutations, driving metabolic signaling pathways such as peroxisome proliferator-activated receptors (PPARs). Understanding the particular vulnerabilities of such cells may uncover novel therapeutic liabilities of the most aggressive cancer cells.
- Published
- 2024
- Full Text
- View/download PDF
10. Na + /H + -exchange inhibition by cariporide is compensated via Na + ,HCO 3 - -cotransport and has no net growth consequences for ErbB2-driven breast carcinomas.
- Author
-
Aaen P, Kristensen KB, Antony A, Hansen SH, Cornett C, Pedersen SF, and Boedtkjer E
- Subjects
- Female, Animals, Mice, Humans, Sodium-Bicarbonate Symporters metabolism, Sodium-Bicarbonate Symporters genetics, Sodium-Bicarbonate Symporters antagonists & inhibitors, Cell Line, Tumor, Hydrogen-Ion Concentration, Guanidines pharmacology, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 genetics, Receptor, ErbB-2 antagonists & inhibitors, Sodium-Hydrogen Exchanger 1 metabolism, Sodium-Hydrogen Exchanger 1 antagonists & inhibitors, Sodium-Hydrogen Exchanger 1 genetics, Sulfones pharmacology, Cell Proliferation drug effects, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Breast Neoplasms metabolism, Breast Neoplasms genetics
- Abstract
Defense against intracellular acidification of breast cancer tissue depends on net acid extrusion via Na
+ ,HCO3 - -cotransporter NBCn1/Slc4a7 and Na+ /H+ -exchanger NHE1/Slc9a1. NBCn1 is increasingly recognized as breast cancer susceptibility protein and promising therapeutic target, whereas evidence for targeting NHE1 is discordant. Currently, selective small molecule inhibitors exist against NHE1 but not NBCn1. Cellular assays-with some discrepancies-link NHE1 activity to proliferation, migration, and invasion; and disrupted NHE1 expression can reduce triple-negative breast cancer growth. Studies on human breast cancer tissue associate high NHE1 expression with reduced metastasis and-in some molecular subtypes-improved patient survival. Here, we evaluate Na+ /H+ -exchange and therapeutic potential of the NHE1 inhibitor cariporide/HOE-642 in murine ErbB2-driven breast cancer. Ex vivo, cariporide inhibits net acid extrusion in breast cancer tissue (IC50 = 0.18 μM) and causes small decreases in steady-state intracellular pH (pHi ). In vivo, we deliver cariporide orally, by osmotic minipumps, and by intra- and peritumoral injections to address the low oral bioavailability and fast metabolism. Prolonged cariporide administration in vivo upregulates NBCn1 expression, shifts pHi regulation towards CO2 /HCO3 - -dependent mechanisms, and shows no net effect on the growth rate of ErbB2-driven primary breast carcinomas. Cariporide also does not influence proliferation markers in breast cancer tissue. Oral, but not parenteral, cariporide elevates serum glucose by ∼1.5 mM. In conclusion, acute administration of cariporide ex vivo powerfully inhibits net acid extrusion from breast cancer tissue but lowers steady-state pHi minimally. Prolonged cariporide administration in vivo is compensated via NBCn1 and we observe no discernible effect on growth of ErbB2-driven breast carcinomas., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Ebbe Boedtkjer reports equipment, drugs, or supplies was provided by Sanofi-Aventis Deutschland GmbH. Kristoffer B. Kristensen reports financial support was provided by Danish Cancer Society. Ebbe Boedtkjer reports financial support was provided by Danish Cancer Society. Ebbe Boedtkjer reports financial support was provided by Novo Nordisk Foundation. Stine F. Pedersen reports a relationship with Solid Therapeutics ApS that includes: board membership and equity or stocks. Ebbe Boedtkjer has patent #EP-3271402 issued to Aarhus University. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
11. Adaptation to an acid microenvironment promotes pancreatic cancer organoid growth and drug resistance.
- Author
-
Stigliani A, Ialchina R, Yao J, Czaplinska D, Dai Y, Andersen HB, Rennie S, Andersson R, Pedersen SF, and Sandelin A
- Subjects
- Animals, Mice, Humans, Hydrogen-Ion Concentration, Acidosis pathology, Acidosis metabolism, Adaptation, Physiological drug effects, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Protein p53 genetics, Cell Survival drug effects, Pancreatic Neoplasms pathology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Organoids drug effects, Organoids metabolism, Organoids pathology, Drug Resistance, Neoplasm genetics, Tumor Microenvironment, Gemcitabine, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Deoxycytidine therapeutic use
- Abstract
Harsh environments in poorly perfused tumor regions may select for traits driving cancer aggressiveness. Here, we investigated whether tumor acidosis interacts with driver mutations to exacerbate cancer hallmarks. We adapted mouse organoids from normal pancreatic duct (mN10) and early pancreatic cancer (mP4, KRAS-G12D mutation, ± p53 knockout) from extracellular pH 7.4 to 6.7, representing acidic niches. Viability was increased by acid adaptation, a pattern most apparent in wild-type (WT) p53 organoids, and exacerbated upon return to pH 7.4. This led to increased survival of acid-adapted organoids treated with gemcitabine and/or erlotinib, and, in WT p53 organoids, acid-induced attenuation of drug effects. New genetic variants became dominant during adaptation, yet they were unlikely to be its main drivers. Transcriptional changes induced by acid and drug adaptation differed overall, but acid adaptation increased the expression of gemcitabine resistance genes. Thus, adaptation to acidosis increases cancer cell viability after chemotherapy., Competing Interests: Declaration of interests S.F.P. is a cofounder of SOLID Therapeutics., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. DLG1 functions upstream of SDCCAG3 and IFT20 to control ciliary targeting of polycystin-2.
- Author
-
Rezi CK, Aslanyan MG, Diwan GD, Cheng T, Chamlali M, Junger K, Anvarian Z, Lorentzen E, Pauly KB, Afshar-Bahadori Y, Fernandes EF, Qian F, Tosi S, Christensen ST, Pedersen SF, Strømgaard K, Russell RB, Miner JH, Mahjoub MR, Boldt K, Roepman R, and Pedersen LB
- Subjects
- Animals, Mice, Humans, Protein Transport, Mice, Knockout, Kidney metabolism, Epithelial Cells metabolism, Protein Binding, Vesico-Ureteral Reflux metabolism, Vesico-Ureteral Reflux genetics, Membrane Proteins metabolism, Membrane Proteins genetics, Urogenital Abnormalities, Cilia metabolism, TRPP Cation Channels metabolism, TRPP Cation Channels genetics, Discs Large Homolog 1 Protein metabolism, Carrier Proteins metabolism, Carrier Proteins genetics
- Abstract
Polarized vesicular trafficking directs specific receptors and ion channels to cilia, but the underlying mechanisms are poorly understood. Here we describe a role for DLG1, a core component of the Scribble polarity complex, in regulating ciliary protein trafficking in kidney epithelial cells. Conditional knockout of Dlg1 in mouse kidney causes ciliary elongation and cystogenesis, and cell-based proximity labeling proteomics and fluorescence microscopy show alterations in the ciliary proteome upon loss of DLG1. Specifically, the retromer-associated protein SDCCAG3, IFT20, and polycystin-2 (PC2) are reduced in the cilia of DLG1-deficient cells compared to control cells. This phenotype is recapitulated in vivo and rescuable by re-expression of wild-type DLG1, but not a Congenital Anomalies of the Kidney and Urinary Tract (CAKUT)-associated DLG1 variant, p.T489R. Finally, biochemical approaches and Alpha Fold modelling suggest that SDCCAG3 and IFT20 form a complex that associates, at least indirectly, with DLG1. Our work identifies a key role for DLG1 in regulating ciliary protein composition and suggests that ciliary dysfunction of the p.T489R DLG1 variant may contribute to CAKUT., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Spatially resolved analysis of microenvironmental gradient impact on cancer cell phenotypes.
- Author
-
Auxillos J, Crouigneau R, Li YF, Dai Y, Stigliani A, Tavernaro I, Resch-Genger U, Sandelin A, Marie R, and Pedersen SF
- Subjects
- Humans, Cell Line, Tumor, Cell Movement, Hydrogen-Ion Concentration, Chemotaxis, Microfluidic Analytical Techniques, Tumor Microenvironment, Phenotype, Neoplasms pathology, Neoplasms metabolism, Neoplasms genetics
- Abstract
Despite the physiological and pathophysiological significance of microenvironmental gradients, e.g., for diseases such as cancer, tools for generating such gradients and analyzing their impact are lacking. Here, we present an integrated microfluidic-based workflow that mimics extracellular pH gradients characteristic of solid tumors while enabling high-resolution live imaging of, e.g., cell motility and chemotaxis, and preserving the capacity to capture the spatial transcriptome. Our microfluidic device generates a pH gradient that can be rapidly controlled to mimic spatiotemporal microenvironmental changes over cancer cells embedded in a 3D matrix. The device can be reopened allowing immunofluorescence analysis of selected phenotypes, as well as the transfer of cells and matrix to a Visium slide for spatially resolved analysis of transcriptional changes across the pH gradient. This workflow is easily adaptable to other gradients and multiple cell types and can therefore prove invaluable for integrated analysis of roles of microenvironmental gradients in biology.
- Published
- 2024
- Full Text
- View/download PDF
14. MCT4 and CD147 colocalize with MMP14 in invadopodia and support matrix degradation and invasion by breast cancer cells.
- Author
-
Meng S, Sørensen EE, Ponniah M, Thorlacius-Ussing J, Crouigneau R, Larsen T, Borre MT, Willumsen N, Flinck M, and Pedersen SF
- Subjects
- Female, Humans, Cell Line, Tumor, Cell Membrane metabolism, Cell Movement, Gelatin metabolism, Lysosomal Membrane Proteins metabolism, Lysosomal Membrane Proteins genetics, Microtubule-Associated Proteins metabolism, Microtubule-Associated Proteins genetics, Muscle Proteins metabolism, Muscle Proteins genetics, Basigin metabolism, Basigin genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Breast Neoplasms genetics, Extracellular Matrix metabolism, Lysosomal-Associated Membrane Protein 1, Matrix Metalloproteinase 14 metabolism, Matrix Metalloproteinase 14 genetics, Monocarboxylic Acid Transporters metabolism, Monocarboxylic Acid Transporters genetics, Neoplasm Invasiveness genetics, Podosomes metabolism
- Abstract
Expression levels of the lactate-H+ cotransporter MCT4 (also known as SLC16A3) and its chaperone CD147 (also known as basigin) are upregulated in breast cancers, correlating with decreased patient survival. Here, we test the hypothesis that MCT4 and CD147 favor breast cancer invasion through interdependent effects on extracellular matrix (ECM) degradation. MCT4 and CD147 expression and membrane localization were found to be strongly reciprocally interdependent in MDA-MB-231 breast cancer cells. Overexpression of MCT4 and/or CD147 increased, and their knockdown decreased, migration, invasion and the degradation of fluorescently labeled gelatin. Overexpression of both proteins led to increases in gelatin degradation and appearance of the matrix metalloproteinase (MMP)-generated collagen-I cleavage product reC1M, and these increases were greater than those observed upon overexpression of each protein alone, suggesting a concerted role in ECM degradation. MCT4 and CD147 colocalized with invadopodia markers at the plasma membrane. They also colocalized with MMP14 and the lysosomal marker LAMP1, as well as partially with the autophagosome marker LC3, in F-actin-decorated intracellular vesicles. We conclude that MCT4 and CD147 reciprocally regulate each other and interdependently support migration and invasiveness of MDA-MB-231 breast cancer cells. Mechanistically, this involves MCT4-CD147-dependent stimulation of ECM degradation and specifically of MMP-mediated collagen-I degradation. We suggest that the MCT4-CD147 complex is co-delivered to invadopodia with MMP14., Competing Interests: Competing interests J.T.-U. and N.W. are employees of Nordic Bioscience A/S, and S.F.P. is cofounder of SOLID Therapeutics. All other authors declare no competing or financial interests., (© 2024. Published by The Company of Biologists Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
15. Transmembrane transporter proteins: Capturing transport in motion.
- Author
-
Loland CJ, Wellendorph P, Pedersen SF, and Gether U
- Subjects
- Carrier Proteins, Biological Transport, Membrane Transport Proteins metabolism, Membrane Proteins
- Published
- 2024
- Full Text
- View/download PDF
16. How protons pave the way to aggressive cancers.
- Author
-
Swietach P, Boedtkjer E, and Pedersen SF
- Subjects
- Humans, Hydrogen-Ion Concentration, Lactic Acid, Tumor Microenvironment physiology, Protons, Neoplasms pathology
- Abstract
Cancers undergo sequential changes to proton (H
+ ) concentration and sensing that are consequences of the disease and facilitate its further progression. The impact of protonation state on protein activity can arise from alterations to amino acids or their titration. Indeed, many cancer-initiating mutations influence pH balance, regulation or sensing in a manner that enables growth and invasion outside normal constraints as part of oncogenic transformation. These cancer-supporting effects become more prominent when tumours develop an acidic microenvironment owing to metabolic reprogramming and disordered perfusion. The ensuing intracellular and extracellular pH disturbances affect multiple aspects of tumour biology, ranging from proliferation to immune surveillance, and can even facilitate further mutagenesis. As a selection pressure, extracellular acidosis accelerates disease progression by favouring acid-resistant cancer cells, which are typically associated with aggressive phenotypes. Although acid-base disturbances in tumours often occur alongside hypoxia and lactate accumulation, there is now ample evidence for a distinct role of H+ -operated responses in key events underpinning cancer. The breadth of these actions presents therapeutic opportunities to change the trajectory of disease., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
17. Lactate receptor GPR81 drives breast cancer growth and invasiveness through regulation of ECM properties and Notch ligand DLL4.
- Author
-
Lundø K, Dmytriyeva O, Spøhr L, Goncalves-Alves E, Yao J, Blasco LP, Trauelsen M, Ponniah M, Severin M, Sandelin A, Kveiborg M, Schwartz TW, and Pedersen SF
- Subjects
- Humans, Female, Lactic Acid metabolism, Ligands, Signal Transduction, Necrosis, Receptor, Notch1 metabolism, Calcium-Binding Proteins genetics, Calcium-Binding Proteins metabolism, Adaptor Proteins, Signal Transducing metabolism, Breast Neoplasms pathology
- 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., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
18. Outcomes After Endovascular Aortic Intervention in Patients With Connective Tissue Disease.
- Author
-
Olsson KW, Mani K, Burdess A, Patterson S, Scali ST, Kölbel T, Panuccio G, Eleshra A, Bertoglio L, Ardita V, Melissano G, Acharya A, Bicknell C, Riga C, Gibbs R, Jenkins M, Bakthavatsalam A, Sweet MP, Kasprzak PM, Pfister K, Oikonomou K, Heloise T, Sobocinski J, Butt T, Dias N, Tang C, Cheng SWK, Vandenhaute S, Van Herzeele I, Sorber RA, Black JH 3rd, Tenorio ER, Oderich GS, Vincent Z, Khashram M, Eagleton MJ, Pedersen SF, Budtz-Lilly J, Lomazzi C, Bissacco D, Trimarchi S, Huerta A, Riambau V, and Wanhainen A
- Subjects
- Humans, Male, Middle Aged, Female, Retrospective Studies, Treatment Outcome, Aorta, Marfan Syndrome complications, Marfan Syndrome surgery, Loeys-Dietz Syndrome complications, Aortic Aneurysm, Thoracic, Endovascular Procedures methods, Connective Tissue Diseases complications, Connective Tissue Diseases surgery, Ehlers-Danlos Syndrome, Type IV
- Abstract
Importance: Endovascular treatment is not recommended for aortic pathologies in patients with connective tissue diseases (CTDs) other than in redo operations and as bridging procedures in emergencies. However, recent developments in endovascular technology may challenge this dogma., Objective: To assess the midterm outcomes of endovascular aortic repair in patients with CTD., Design, Setting, and Participants: For this descriptive retrospective study, data on demographics, interventions, and short-term and midterm outcomes were collected from 18 aortic centers in Europe, Asia, North America, and New Zealand. Patients with CTD who had undergone endovascular aortic repair from 2005 to 2020 were included. Data were analyzed from December 2021 to November 2022., Exposure: All principal endovascular aortic repairs, including redo surgery and complex repairs of the aortic arch and visceral aorta., Main Outcomes and Measures: Short-term and midterm survival, rates of secondary procedures, and conversion to open repair., Results: In total, 171 patients were included: 142 with Marfan syndrome, 17 with Loeys-Dietz syndrome, and 12 with vascular Ehlers-Danlos syndrome (vEDS). Median (IQR) age was 49.9 years (37.9-59.0), and 107 patients (62.6%) were male. One hundred fifty-two (88.9%) were treated for aortic dissections and 19 (11.1%) for degenerative aneurysms. One hundred thirty-six patients (79.5%) had undergone open aortic surgery before the index endovascular repair. In 74 patients (43.3%), arch and/or visceral branches were included in the repair. Primary technical success was achieved in 168 patients (98.2%), and 30-day mortality was 2.9% (5 patients). Survival at 1 and 5 years was 96.2% and 80.6% for Marfan syndrome, 93.8% and 85.2% for Loeys-Dietz syndrome, and 75.0% and 43.8% for vEDS, respectively. After a median (IQR) follow-up of 4.7 years (1.9-9.2), 91 patients (53.2%) had undergone secondary procedures, of which 14 (8.2%) were open conversions., Conclusions and Relevance: This study found that endovascular aortic interventions, including redo procedures and complex repairs of the aortic arch and visceral aorta, in patients with CTD had a high rate of early technical success, low perioperative mortality, and a midterm survival rate comparable with reports of open aortic surgery in patients with CTD. The rate of secondary procedures was high, but few patients required conversion to open repair. Improvements in devices and techniques, as well as ongoing follow-up, may result in endovascular treatment for patients with CTD being included in guideline recommendations.
- Published
- 2023
- Full Text
- View/download PDF
19. Imaging of atherosclerosis with [ 64 Cu]Cu-DOTA-TATE in a translational head-to-head comparison study with [ 18 F]FDG, and Na[ 18 F]F in rabbits.
- Author
-
Grandjean CE, Pedersen SF, Christensen C, Dibenedetto A, Eriksen T, Binderup T, and Kjaer A
- Subjects
- Rabbits, Male, Animals, Tomography, X-Ray Computed, Positron-Emission Tomography methods, Positron Emission Tomography Computed Tomography, Inflammation pathology, Fluorodeoxyglucose F18, Atherosclerosis diagnostic imaging, Atherosclerosis pathology
- Abstract
Atherosclerosis is a chronic inflammatory disease of the larger arteries that may lead to cardiovascular events. Identification of patients at highest risk of cardiovascular events is challenging, but molecular imaging using positron emission tomography (PET) may prove useful. The aim of this study was to evaluate and compare head-to-head three different PET tracers. Furthermore, tracer uptake is compared to gene expression alterations of the arterial vessel wall. Male New Zealand White rabbits (control group; n = 10, atherosclerotic group; n = 11) were used for the study. Vessel wall uptake was assessed with the three different PET tracers: [
18 F]FDG (inflammation), Na[18 F]F (microcalcification), and [64 Cu]Cu-DOTA-TATE (macrophages), using PET/computed tomography (CT). Tracer uptake was measured as standardized uptake value (SUV), and arteries from both groups were analyzed ex vivo by autoradiography, qPCR, histology, and immunohistochemistry. In rabbits, the atherosclerotic group showed significantly higher uptake of all three tracers compared to the control group [18 F]FDG: SUVmean 1.50 ± 0.11 versus 1.23 ± 0.09, p = 0.025; Na[18 F]F: SUVmean 1.54 ± 0.06 versus 1.18 ± 0.10, p = 0.006; and [64 Cu]Cu-DOTA-TATE: SUVmean 2.30 ± 0.27 versus 1.65 ± 0.16; p = 0.047. Of the 102 genes analyzed, 52 were differentially expressed in the atherosclerotic group compared to the control group and several genes correlated with tracer uptake. In conclusion, we demonstrated the diagnostic value of [64 Cu]Cu-DOTA-TATE and Na[18 F]F for identifying atherosclerosis in rabbits. The two PET tracers provided information distinct from that obtained with [18 F]FDG. None of the three tracers correlated significantly to each other, but [64 Cu]Cu-DOTA-TATE and Na[18 F]F uptake both correlated with markers of inflammation. [64 Cu]Cu-DOTA-TATE was higher in atherosclerotic rabbits compared to [18 F]FDG and Na[18 F]F., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
20. The prolactin receptor scaffolds Janus kinase 2 via co-structure formation with phosphoinositide-4,5-bisphosphate.
- Author
-
Araya-Secchi R, Bugge K, Seiffert P, Petry A, Haxholm GW, Lindorff-Larsen K, Pedersen SF, Arleth L, and Kragelund BB
- Subjects
- Humans, Carrier Proteins metabolism, Phosphorylation, Prolactin metabolism, Signal Transduction, STAT5 Transcription Factor metabolism, Janus Kinase 2 metabolism, Receptors, Prolactin metabolism
- Abstract
Class 1 cytokine receptors transmit signals through the membrane by a single transmembrane helix to an intrinsically disordered cytoplasmic domain that lacks kinase activity. While specific binding to phosphoinositides has been reported for the prolactin receptor (PRLR), the role of lipids in PRLR signaling is unclear. Using an integrative approach combining nuclear magnetic resonance spectroscopy, cellular signaling experiments, computational modeling, and simulation, we demonstrate co-structure formation of the disordered intracellular domain of the human PRLR, the membrane constituent phosphoinositide-4,5-bisphosphate (PI(4,5)P
2 ) and the FERM-SH2 domain of the Janus kinase 2 (JAK2). We find that the complex leads to accumulation of PI(4,5)P2 at the transmembrane helix interface and that the mutation of residues identified to interact specifically with PI(4,5)P2 negatively affects PRLR-mediated activation of signal transducer and activator of transcription 5 (STAT5). Facilitated by co-structure formation, the membrane-proximal disordered region arranges into an extended structure. We suggest that the co-structure formed between PRLR, JAK2, and PI(4,5)P2 locks the juxtamembrane disordered domain of the PRLR in an extended structure, enabling signal relay from the extracellular to the intracellular domain upon ligand binding. We find that the co-structure exists in different states which we speculate could be relevant for turning signaling on and off. Similar co-structures may be relevant for other non-receptor tyrosine kinases and their receptors., Competing Interests: RA, KB, PS, AP, GH, KL, SP, LA, BK No competing interests declared, (© 2023, Araya-Secchi et al.)- Published
- 2023
- Full Text
- View/download PDF
21. Efficacy and Safety of Exercise Testing and Rehabilitation for Aortic Dissection Patients: A SCOPING REVIEW.
- Author
-
Jepsen LR, D'Oria M, Pedersen SF, and Budtz-Lilly J
- Subjects
- Humans, Exercise, Exercise Test, Exercise Therapy, Aortic Dissection, Quality of Life
- Abstract
Objective: Although physical exercise has established benefits for long-term cardiovascular health, concern regarding further aortic events has limited the evidence for exercise among aortic dissection (AD) patients. The objective was to perform a scoping review of the current concepts and gaps in the literature regarding the benefit and safety of cardiovascular testing and rehabilitation among post-AD patients., Review Methods: A scoping review of the literature search was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR). Eligibility criteria included any studies with AD in relation to exercise rehabilitation or testing and physical or mental health. Electronic databases were queried for relevant studies (last queried, November 1, 2021)., Results: Six observational studies were included with a total of 381 patients with AD, all of whom underwent surgical intervention. Study heterogeneity prevented data synthesis and a formal systematic review, although four dominant themes emerged: cardiovascular-related outcomes, aorta-related outcomes, quality of life (QoL), and serious adverse events. Patients with AD have reduced baseline physical capacity and QoL. Rehabilitation programs may increase both physical status and QoL. Rates of serious adverse rates are minimal yet poorly defined., Conclusions: Within the context of apparent benefits from cardiovascular testing and rehabilitation for patients with AD who have undergone intervention, this scoping review highlights the need for increased comparative research specific to exercise among patients with AD and outcomes such as mortality and reinterventions., Competing Interests: The authors declare no conflicts of interest., (Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. Chronic acidosis rewires cancer cell metabolism through PPARα signaling.
- Author
-
Rolver MG, Holland LKK, Ponniah M, Prasad NS, Yao J, Schnipper J, Kramer S, Elingaard-Larsen L, Pedraz-Cuesta E, Liu B, Pardo LA, Maeda K, Sandelin A, and Pedersen SF
- Subjects
- Humans, Transcription Factors genetics, Gene Expression Regulation, PPAR alpha genetics, PPAR alpha metabolism, Fatty Acids metabolism, Lipid Metabolism, Liver metabolism, Tumor Microenvironment, Neoplasms metabolism, Acidosis
- Abstract
The mechanisms linking tumor microenvironment acidosis to disease progression are not understood. Here, we used mammary, pancreatic, and colon cancer cells to show that adaptation to growth at an extracellular pH (pH
e ) mimicking acidic tumor niches is associated with upregulated net acid extrusion capacity and elevated intracellular pH at physiological pHe , but not at acidic pHe . Using metabolic profiling, shotgun lipidomics, imaging and biochemical analyses, we show that the acid adaptation-induced phenotype is characterized by a shift toward oxidative metabolism, increased lipid droplet-, triacylglycerol-, peroxisome content and mitochondrial hyperfusion. Peroxisome proliferator-activated receptor-α (PPARA, PPARα) expression and activity are upregulated, at least in part by increased fatty acid uptake. PPARα upregulates genes driving increased mitochondrial and peroxisomal mass and β-oxidation capacity, including mitochondrial lipid import proteins CPT1A, CPT2 and SLC25A20, electron transport chain components, peroxisomal proteins PEX11A and ACOX1, and thioredoxin-interacting protein (TXNIP), a negative regulator of glycolysis. This endows acid-adapted cancer cells with increased capacity for utilizing fatty acids for metabolic needs, while limiting glycolysis. As a consequence, the acid-adapted cells exhibit increased sensitivity to PPARα inhibition. We conclude that PPARα is a key upstream regulator of metabolic changes favoring cancer cell survival in acidic tumor niches., (© 2022 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)- Published
- 2023
- Full Text
- View/download PDF
23. Dynamic localization of the Na+-HCO3- co-transporter NBCn1 to the plasma membrane, centrosomes, spindle and primary cilia.
- Author
-
Severin M, Pedersen EL, Borre MT, Axholm I, Christiansen FB, Ponniah M, Czaplinska D, Larsen T, Pardo LA, and Pedersen SF
- Subjects
- Humans, Animals, Rats, Cell Cycle, Cyclic AMP metabolism, Cell Polarity, Epithelial Cells metabolism, Cell Membrane chemistry, Cilia chemistry, Centrosome chemistry, Spindle Apparatus chemistry, Sodium-Bicarbonate Symporters analysis, Sodium-Bicarbonate Symporters metabolism
- Abstract
Finely tuned regulation of transport protein localization is vital for epithelial function. The Na+-HCO3- co-transporter NBCn1 (also known as SLC4A7) is a key contributor to epithelial pH homeostasis, yet the regulation of its subcellular localization is not understood. Here, we show that a predicted N-terminal β-sheet and short C-terminal α-helical motif are essential for NBCn1 plasma membrane localization in epithelial cells. This localization was abolished by cell-cell contact disruption, and co-immunoprecipitation (co-IP) and proximity ligation (PLA) revealed NBCn1 interaction with E-cadherin and DLG1, linking it to adherens junctions and the Scribble complex. NBCn1 also interacted with RhoA and localized to lamellipodia and filopodia in migrating cells. Finally, analysis of native and GFP-tagged NBCn1 localization, subcellular fractionation, co-IP with Arl13B and CEP164, and PLA of NBCn1 and tubulin in mitotic spindles led to the surprising conclusion that NBCn1 additionally localizes to centrosomes and primary cilia in non-dividing, polarized epithelial cells, and to the spindle, centrosomes and midbodies during mitosis. We propose that NBCn1 traffics between lateral junctions, the leading edge and cell division machinery in Rab11 endosomes, adding new insight to the role of NBCn1 in cell cycle progression., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
24. Crosstalk between tumor acidosis, p53 and extracellular matrix regulates pancreatic cancer aggressiveness.
- Author
-
Czaplinska D, Ialchina R, Andersen HB, Yao J, Stigliani A, Dannesboe J, Flinck M, Chen X, Mitrega J, Gnosa SP, Dmytriyeva O, Alves F, Napp J, Sandelin A, and Pedersen SF
- Subjects
- Humans, Cell Line, Tumor, Extracellular Matrix metabolism, Proto-Oncogene Proteins c-akt metabolism, Transforming Growth Factor beta metabolism, Tumor Microenvironment, Tumor Suppressor Protein p53 genetics, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy with minimal treatment options and a global rise in prevalence. PDAC is characterized by frequent driver mutations including KRAS and TP53 (p53), and a dense, acidic tumor microenvironment (TME). The relation between genotype and TME in PDAC development is unknown. Strikingly, when wild type (WT) Panc02 PDAC cells were adapted to growth in an acidic TME and returned to normal pH to mimic invasive cells escaping acidic regions, they displayed a strong increase of aggressive traits such as increased growth in 3-dimensional (3D) culture, adhesion-independent colony formation and invasive outgrowth. This pattern of acidosis-induced aggressiveness was observed in 3D spheroid culture as well as upon organotypic growth in matrigel, collagen-I and combination thereof, mimicking early and later stages of PDAC development. Acid-adaptation-induced gain of cancerous traits was further increased by p53 knockout (KO), but only in specific extracellular matrix (ECM) compositions. Akt- and Transforming growth factor-β (TGFβ) signaling, as well as expression of the Na
+ /H+ exchanger NHE1, were increased by acid adaptation. Whereas Akt inhibition decreased spheroid growth regardless of treatment and genotype, stimulation with TGFβI increased growth of WT control spheroids, and inhibition of TGFβ signaling tended to limit growth under acidic conditions only. Our results indicate that a complex crosstalk between tumor acidosis, ECM composition and genotype contributes to PDAC development. The findings may guide future strategies for acidosis-targeted therapies., (© 2022 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)- Published
- 2023
- Full Text
- View/download PDF
25. Aquaporin water channels affect the response of conventional anticancer therapies of 3D grown breast cancer cells.
- Author
-
Edamana S, Pedersen SF, and Nejsum LN
- Subjects
- Humans, Female, Fluorouracil pharmacology, Doxorubicin pharmacology, Aquaporin 1 genetics, Aquaporin 1 metabolism, Aquaporin 5 metabolism, Aquaporin 3 genetics, Aquaporin 3 metabolism, Aquaporin 4, Aquaporin 2, Breast Neoplasms drug therapy, Aquaporins metabolism
- Abstract
Aquaporin (AQP) water channels facilitate water transport across cellular membranes and are essential in regulation of body water balance. Moreover, several AQPs are overexpressed or ectopically expressed in breast cancer. Interestingly, several in vitro studies have suggested that AQPs can affect the response to conventional anticancer chemotherapies. Therefore, we took a systematic approach to test how AQP1, AQP3 and AQP5, which are often over-/ectopically expressed in breast cancer, affect total viability of 3-dimensional (3D) breast cancer cell spheroids when treated with the conventional anticancer chemotherapies Cisplatin, 5-Fluorouracil (5-FU) and Doxorubicin, a Combination of the three drugs as well as the Combination plus the Ras inhibitor Salirasib. Total viability of spheroids overexpressing AQP1 were decreased by all treatments except for 5-FU, which increased total viability by 20% compared to DMSO treated controls. All treatments reduced viability of spheroids overexpressing AQP3. In contrast, only Doxorubicin, Combination and Combination + Salirasib reduced total viability of spheroids overexpressing AQP5. Thus, this study supports a significant role of AQPs in the response to conventional chemotherapies. Evaluating the role of individual proteins that contribute to resistance to chemotherapies is essential in advancing personalized medicine in breast carcinomas., Competing Interests: Declaration of competing interest The authors declare no conflict of interest., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Acid Adaptation Promotes TRPC1 Plasma Membrane Localization Leading to Pancreatic Ductal Adenocarcinoma Cell Proliferation and Migration through Ca 2+ Entry and Interaction with PI3K/CaM.
- Author
-
Schnipper J, Kouba S, Hague F, Girault A, Telliez MS, Guénin S, Ahidouch A, Pedersen SF, and Ouadid-Ahidouch H
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a low overall survival rate of less than 10% and limited therapeutic options. Fluctuations in tumor microenvironment pH are a hallmark of PDAC development and progression. Many ion channels are bona fide cellular sensors of changes in pH. Yet, the interplay between the acidic tumor microenvironment and ion channel regulation in PDAC is poorly understood. In this study, we show that acid adaption increases PANC-1 cell migration but attenuates proliferation and spheroid growth, which are restored upon recovery. Moreover, acid adaptation and recovery conditions favor the plasma membrane localization of the pH-sensitive calcium (Ca
2+ ) channel transient receptor potential C1 (TRPC1), TRPC1-mediated Ca2+ influx, channel interaction with the PI3K p85α subunit and calmodulin (CaM), and AKT and ERK1/2 activation. Knockdown (KD) of TRPC1 suppresses cell migration, proliferation, and spheroid growth, notably in acid-recovered cells. KD of TRPC1 causes the accumulation of cells in G0/G1 and G2/M phases, along with reduced expression of CDK6, -2, and -1, and cyclin A, and increased expression of p21CIP1 . TRPC1 silencing decreases the basal Ca2+ influx in acid-adapted and -recovered cells, but not in normal pH conditions, and Ca2+ chelation reduces cell migration and proliferation solely in acid adaptation and recovery conditions. In conclusion, acid adaptation and recovery reinforce the involvement of TRPC1 in migration, proliferation, and cell cycle progression by permitting Ca2+ entry and forming a complex with the PI3K p85α subunit and CaM.- Published
- 2022
- Full Text
- View/download PDF
27. The TRPC1 Channel Forms a PI3K/CaM Complex and Regulates Pancreatic Ductal Adenocarcinoma Cell Proliferation in a Ca 2+ -Independent Manner.
- Author
-
Schnipper J, Kouba S, Hague F, Girault A, Rybarczyk P, Telliez MS, Guénin S, Tebbakha R, Sevestre H, Ahidouch A, Pedersen SF, and Ouadid-Ahidouch H
- Subjects
- Calcium metabolism, Calmodulin metabolism, Cell Proliferation, Humans, Phosphatidylinositol 3-Kinases metabolism, TRPC Cation Channels genetics, TRPC Cation Channels metabolism, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal genetics, Pancreatic Neoplasms genetics
- Abstract
Dysregulation of the transient receptor canonical ion channel (TRPC1) has been found in several cancer types, yet the underlying molecular mechanisms through which TRPC1 impacts pancreatic ductal adenocarcinoma (PDAC) cell proliferation are incompletely understood. Here, we found that TRPC1 is upregulated in human PDAC tissue compared to adjacent pancreatic tissue and this higher expression correlates with low overall survival. TRPC1 is, as well, upregulated in the aggressive PDAC cell line PANC-1, compared to a duct-like cell line, and its knockdown (KD) reduced cell proliferation along with PANC-1 3D spheroid growth by arresting cells in the G1/S phase whilst decreasing cyclin A, CDK2, CDK6, and increasing p21
CIP1 expression. In addition, the KD of TRPC1 neither affected Ca2+ influx nor store-operated Ca2+ entry (SOCE) and reduced cell proliferation independently of extracellular calcium. Interestingly, TRPC1 interacted with the PI3K-p85α subunit and calmodulin (CaM); both the CaM protein level and AKT phosphorylation were reduced upon TRPC1 KD. In conclusion, our results show that TRPC1 regulates PDAC cell proliferation and cell cycle progression by interacting with PI3K-p85α and CaM through a Ca2+ -independent pathway.- Published
- 2022
- Full Text
- View/download PDF
28. Inhibition of a Chromatin and Transcription Modulator, SLTM, Increases HIV-1 Reactivation Identified by a CRISPR Inhibition Screen.
- Author
-
Pedersen SF, Collora JA, Kim RN, Yang K, Razmi A, Catalano AA, Yeh YJ, Mounzer K, Tebas P, Montaner LJ, and Ho YC
- Subjects
- Antiretroviral Therapy, Highly Active, CD4-Positive T-Lymphocytes, Chromatin genetics, Chromatin metabolism, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Gene Knockdown Techniques, Humans, Jurkat Cells, Matrix Attachment Region Binding Proteins antagonists & inhibitors, Matrix Attachment Region Binding Proteins metabolism, HIV Infections physiopathology, HIV Seropositivity genetics, HIV-1 physiology, Virus Activation genetics
- Abstract
Despite effective antiretroviral therapy, HIV-1 persistence in latent reservoirs remains a major obstacle to a cure. We postulate that HIV-1 silencing factors suppress HIV-1 reactivation and that inhibition of these factors will increase HIV-1 reactivation. To identify HIV-1 silencing factors, we conducted a genome-wide CRISPR inhibition (CRISPRi) screen using four CRISPRi-ready, HIV-1-d6-GFP-infected Jurkat T cell clones with distinct integration sites. We sorted cells with increased green fluorescent protein (GFP) expression and captured single guide RNAs (sgRNAs) via targeted deep sequencing. We identified 18 HIV-1 silencing factors that were significantly enriched in HIV-1-d6-GFP
high cells. Among them, SLTM (scaffold attachment factor B-like transcription modulator) is an epigenetic and transcriptional modulator having both DNA and RNA binding capacities not previously known to affect HIV-1 transcription. Knocking down SLTM by CRISPRi significantly increased HIV-1-d6-GFP expression (by 1.9- to 4.2-fold) in three HIV-1-d6-GFP-Jurkat T cell clones. Furthermore, SLTM knockdown increased the chromatin accessibility of HIV-1 and the gene in which HIV-1 is integrated but not the housekeeping gene POLR2A . To test whether SLTM inhibition can reactivate HIV-1 and further induce cell death of HIV-1-infected cells ex vivo , we established a small interfering RNA (siRNA) knockdown method that reduced SLTM expression in CD4+ T cells from 10 antiretroviral therapy (ART)-treated, virally suppressed, HIV-1-infected individuals ex vivo . Using limiting dilution culture, we found that SLTM knockdown significantly reduced the frequency of HIV-1-infected cells harboring inducible HIV-1 by 62.2% (0.56/106 versus 1.48/106 CD4+ T cells [ P = 0.029]). Overall, our study indicates that SLTM inhibition reactivates HIV-1 in vitro and induces cell death of HIV-1-infected cells ex vivo . Our study identified SLTM as a novel therapeutic target. IMPORTANCE HIV-1-infected cells, which can survive drug treatment and immune cell killing, prevent an HIV-1 cure. Immune recognition of infected cells requires HIV-1 protein expression; however, HIV-1 protein expression is limited in infected cells after long-term therapy. The ways in which the HIV-1 provirus is blocked from producing protein are unknown. We identified a new host protein that regulates HIV-1 gene expression. We also provided a new method of studying HIV-1-host factor interactions in cells from infected individuals. These improvements may enable future strategies to reactivate HIV-1 in infected individuals so that infected cells can be killed by immune cells, drug treatment, or the virus itself.- Published
- 2022
- Full Text
- View/download PDF
29. In vivo detection of urokinase-type plasminogen activator receptor (uPAR) expression in arterial atherogenesis using [ 64 Cu]Cu-DOTA-AE105 positron emission tomography (PET).
- Author
-
Khare HA, Døssing KBV, Ringgaard L, Christensen E, Urbak L, Sillesen H, Ripa RS, Binderup T, Pedersen SF, and Kjaer A
- Subjects
- Arteries metabolism, Copper Radioisotopes, Heterocyclic Compounds, 1-Ring, Humans, Oligopeptides, Positron Emission Tomography Computed Tomography, Positron-Emission Tomography methods, Receptors, Urokinase Plasminogen Activator genetics, Receptors, Urokinase Plasminogen Activator metabolism, Retrospective Studies, Urokinase-Type Plasminogen Activator, Atherosclerosis diagnostic imaging, Atherosclerosis genetics, Plaque, Atherosclerotic
- Abstract
Background and Aims: Urokinase-type plasminogen activator receptor (uPAR) is associated with extracellular matrix (ECM) degradation and cancer aggressiveness. Its role in arterial atherogenesis as a molecular imaging target is not well-established. The aim of this study was to non-invasively visualize uPAR expression in atherosclerosis by a novel uPAR-targeting positron emission tomography (PET) tracer [
64 Cu]Cu-DOTA-AE105., Methods: We used molecular biology to investigate uPAR expression by analyzing human atherosclerotic plaques and cultured cells. A retrospective analysis was performed on patients, who underwent combined PET/CT (n = 10) to measure [64 Cu]Cu-DOTA-AE105 uptake in five large arteries, divided into a high and low-risk group based on coronary artery calcium score (CAC score)., Results: The in vitro assay for THP-1 monocytes displayed a significantly upregulated uPAR expression upon stimulation (5.2-fold upregulation, p < 0.0001 by a one-way ANOVA followed by Tukey's test) by single-cell flowcytometric analysis. Freshly excised human atherosclerotic plaques underwent flow cytometric and microarray analyses manifesting 73.9 ± 2.9% of mononuclear phagocyte system (MPS) cells expressing uPAR and had a greater than 7-fold higher gene expression of plasminogen activator urokinase receptor (PLAUR, p = 0.002), integrin subunit alpha X (ITGAX, p = 0.0008), and cluster of differentiation 163 (CD163, p < 0.0001). The tissue-to-background ratios (TBRmax ) in five large arteries showed a higher [64 Cu]Cu-DOTA-AE105 uptake in the group with high CAC score compared to the group with low CAC score (2.4 ± 0.1 vs 1.7 ± 0.1, p = 0.057), significantly higher in the ascending aorta (2.7 ± 0.1 vs 2.0 ± 0.1, p = 0.038) and the abdominal aorta (3.2 ± 0.2 vs 2.0 ± 0.2, p = 0.038) by a non-parametric Mann-Whitney test., Conclusions: uPAR is abundantly expressed by MPS cells in atherosclerotic plaques and can be visualized by the novel PET tracer [64 Cu]Cu-DOTA-AE105 that may non-invasively detect extracellular matrix remodeling during atherogenesis., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
30. How Reciprocal Interactions Between the Tumor Microenvironment and Ion Transport Proteins Drive Cancer Progression.
- Author
-
Elingaard-Larsen LO, Rolver MG, Sørensen EE, and Pedersen SF
- Subjects
- Carrier Proteins therapeutic use, Endothelial Cells, Humans, Neoplastic Processes, Neoplasms drug therapy, Tumor Microenvironment
- Abstract
Solid tumors comprise two major components: the cancer cells and the tumor stroma. The stroma is a mixture of cellular and acellular components including fibroblasts, mesenchymal and cancer stem cells, endothelial cells, immune cells, extracellular matrix, and tumor interstitial fluid. The insufficient tumor perfusion and the highly proliferative state and dysregulated metabolism of the cancer cells collectively create a physicochemical microenvironment characterized by altered nutrient concentrations and varying degrees of hypoxia and acidosis. Furthermore, both cancer and stromal cells secrete numerous growth factors, cytokines, and extracellular matrix proteins which further shape the tumor microenvironment (TME), favoring cancer progression.Transport proteins expressed by cancer and stromal cells localize at the interface between the cells and the TME and are in a reciprocal relationship with it, as both sensors and modulators of TME properties. It has been amply demonstrated how acid-base and nutrient transporters of cancer cells enable their growth, presumably by contributing both to the extracellular acidosis and the exchange of metabolic substrates and waste products between cells and TME. However, the TME also impacts other transport proteins important for cancer progression, such as multidrug resistance proteins. In this review, we summarize current knowledge of the cellular and acellular components of solid tumors and their interrelationship with key ion transport proteins. We focus in particular on acid-base transport proteins with known or proposed roles in cancer development, and we discuss their relevance for novel therapeutic strategies., (© 2020. Springer Nature Switzerland AG.)
- Published
- 2022
- Full Text
- View/download PDF
31. [Stenting of uncomplicated type B aortic dissection].
- Author
-
Mildahl B, Budtz-Lilly J, Mafi HM, Vammen S, and Pedersen SF
- Subjects
- Humans, Retrospective Studies, Risk Factors, Treatment Outcome, Aortic Dissection diagnostic imaging, Aortic Dissection surgery, Aortic Aneurysm, Thoracic diagnostic imaging, Aortic Aneurysm, Thoracic surgery, Blood Vessel Prosthesis Implantation, Endovascular Procedures
- Abstract
Conservative treatment of uncomplicated type B aortic dissection is associated with low short-term mortality. However long-term mortality and complication rates are high, suggesting the need for more aggressive treatment. Studies suggest that combining medical treatment with thoracic endovascular aortic repair (TEVAR) is associated with a better prognosis. TEVAR is not without risk, however, and it is paramount to identify characteristics of risk and benefit as summarised in this review. Further studies are needed in order to implement improved patient selection.
- Published
- 2021
32. Metabolic reprogramming by driver mutation-tumor microenvironment interplay in pancreatic cancer: new therapeutic targets.
- Author
-
Andersen HB, Ialchina R, Pedersen SF, and Czaplinska D
- Subjects
- Humans, Mutation, Tumor Microenvironment genetics, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers globally with a mortality rate exceeding 95% and very limited therapeutic options. A hallmark of PDAC is its acidic tumor microenvironment, further characterized by excessive fibrosis and depletion of oxygen and nutrients due to poor vascularity. The combination of PDAC driver mutations and adaptation to this hostile environment drives extensive metabolic reprogramming of the cancer cells toward non-canonical metabolic pathways and increases reliance on scavenging mechanisms such as autophagy and macropinocytosis. In addition, the cancer cells benefit from metabolic crosstalk with nonmalignant cells within the tumor microenvironment, including pancreatic stellate cells, fibroblasts, and endothelial and immune cells. Increasing evidence shows that this metabolic rewiring is closely related to chemo- and radioresistance and immunosuppression, causing extensive treatment failure. Indeed, stratification of human PDAC tumors into subtypes based on their metabolic profiles was shown to predict disease outcome. Accordingly, an increasing number of clinical trials target pro-tumorigenic metabolic pathways, either as stand-alone treatment or in conjunction with chemotherapy. In this review, we highlight key findings and potential future directions of pancreatic cancer metabolism research, specifically focusing on novel therapeutic opportunities., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
33. The Voltage-Gated Sodium Channel Beta4 Subunit Maintains Epithelial Phenotype in Mammary Cells.
- Author
-
Doray A, Lemoine R, Severin M, Chadet S, Lopez-Charcas O, Héraud A, Baron C, Besson P, Monteil A, Pedersen SF, and Roger S
- Subjects
- Animals, Cell Line, Cell Polarity, Down-Regulation, Epithelial Cells cytology, Female, Humans, Mesoderm metabolism, Phenotype, Proteolysis, beta Catenin metabolism, Epithelial Cells metabolism, Mammary Glands, Animal cytology, Protein Subunits metabolism, Voltage-Gated Sodium Channel beta-4 Subunit metabolism
- Abstract
The SCN4B gene, coding for the Na
V β4 subunit of voltage-gated sodium channels, was recently found to be expressed in normal epithelial cells and down-regulated in several cancers. However, its function in normal epithelial cells has not been characterized. In this study, we demonstrated that reducing NaV β4 expression in MCF10A non-cancer mammary epithelial cells generated important morphological changes observed both in two-dimensional cultures and in three-dimensional cysts. Most notably, the loss of NaV β4 induced a complete loss of epithelial organisation in cysts and increased proteolytic activity towards the extracellular matrix. Loss of epithelial morphology was associated with an increased degradation of β-catenin, reduced E-cadherin expression and induction of mesenchymal markers N-cadherin, vimentin, and α-SMA expression. Overall, our results suggest that Navβ4 may participate in the maintenance of the epithelial phenotype in mammary cells and that its downregulation might be a determining step in early carcinogenesis.- Published
- 2021
- Full Text
- View/download PDF
34. Restriction of SARS-CoV-2 replication by targeting programmed -1 ribosomal frameshifting.
- Author
-
Sun Y, Abriola L, Niederer RO, Pedersen SF, Alfajaro MM, Silva Monteiro V, Wilen CB, Ho YC, Gilbert WV, Surovtseva YV, Lindenbach BD, and Guo JU
- Subjects
- Animals, Betacoronavirus, Chlorocebus aethiops, Fluoroquinolones pharmacology, Frameshifting, Ribosomal genetics, Mutation, Nucleic Acid Conformation, RNA, Viral chemistry, RNA, Viral genetics, SARS-CoV-2 physiology, Vero Cells, Antiviral Agents pharmacology, Frameshifting, Ribosomal drug effects, SARS-CoV-2 drug effects, Virus Replication drug effects
- Abstract
Translation of open reading frame 1b (ORF1b) in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires a programmed -1 ribosomal frameshift (-1 PRF) promoted by an RNA pseudoknot. The extent to which SARS-CoV-2 replication may be sensitive to changes in -1 PRF efficiency is currently unknown. Through an unbiased, reporter-based high-throughput compound screen, we identified merafloxacin, a fluoroquinolone antibacterial, as a -1 PRF inhibitor for SARS-CoV-2. Frameshift inhibition by merafloxacin is robust to mutations within the pseudoknot region and is similarly effective on -1 PRF of other betacoronaviruses. Consistent with the essential role of -1 PRF in viral gene expression, merafloxacin impedes SARS-CoV-2 replication in Vero E6 cells, thereby providing proof-of-principle for targeting -1 PRF as a plausible and effective antiviral strategy for SARS-CoV-2 and other coronaviruses., Competing Interests: Competing interest statement: Yale University has filed a provisional patent application related to this work titled “Compounds and Compositions for Disrupting Programmed Ribosomal Frameshifting.”, (Copyright © 2021 the Author(s). Published by PNAS.)
- Published
- 2021
- Full Text
- View/download PDF
35. The Interplay between Dysregulated Ion Transport and Mitochondrial Architecture as a Dangerous Liaison in Cancer.
- Author
-
Pedersen SF, Flinck M, and Pardo LA
- Subjects
- Calcium metabolism, Cell Movement, Cell Proliferation, Homeostasis, Humans, Ion Channels metabolism, Neoplastic Stem Cells metabolism, Potassium metabolism, Protons, Tumor Microenvironment, Ion Transport, Mitochondria metabolism, Neoplasms metabolism, Neoplasms pathology
- Abstract
Transport of ions and nutrients is a core mitochondrial function, without which there would be no mitochondrial metabolism and ATP production. Both ion homeostasis and mitochondrial phenotype undergo pervasive changes during cancer development, and both play key roles in driving the malignancy. However, the link between these events has been largely ignored. This review comprehensively summarizes and critically discusses the role of the reciprocal relationship between ion transport and mitochondria in crucial cellular functions, including metabolism, signaling, and cell fate decisions. We focus on Ca
2+ , H+ , and K+ , which play essential and highly interconnected roles in mitochondrial function and are profoundly dysregulated in cancer. We describe the transport and roles of these ions in normal mitochondria, summarize the changes occurring during cancer development, and discuss how they might impact tumorigenesis.- Published
- 2021
- Full Text
- View/download PDF
36. Dynamic Na + /H + exchanger 1 (NHE1) - calmodulin complexes of varying stoichiometry and structure regulate Ca 2+ -dependent NHE1 activation.
- Author
-
Sjøgaard-Frich LM, Prestel A, Pedersen ES, Severin M, Kristensen KK, Olsen JG, Kragelund BB, and Pedersen SF
- Subjects
- Calmodulin metabolism, Calorimetry, Cell Line, Cytosol metabolism, Fibroblasts, Humans, Magnetic Resonance Spectroscopy, Sodium-Hydrogen Exchanger 1 metabolism, Calcium metabolism, Calmodulin genetics, Sodium-Hydrogen Exchanger 1 genetics
- Abstract
Calmodulin (CaM) engages in Ca
2+ -dependent interactions with numerous proteins, including a still incompletely understood physical and functional interaction with the human Na+ /H+ -exchanger NHE1. Using nuclear magnetic resonance (NMR) spectroscopy, isothermal titration calorimetry, and fibroblasts stably expressing wildtype and mutant NHE1, we discovered multiple accessible states of this functionally important complex existing in different NHE1:CaM stoichiometries and structures. We determined the NMR solution structure of a ternary complex in which CaM links two NHE1 cytosolic tails. In vitro , stoichiometries and affinities could be tuned by variations in NHE1:CaM ratio and calcium ([Ca2+ ]) and by phosphorylation of S648 in the first CaM-binding α-helix. In cells, Ca2+ -CaM-induced NHE1 activity was reduced by mimicking S648 phosphorylation and by mutation of the first CaM-binding α-helix, whereas it was unaffected by inhibition of Akt, one of several kinases phosphorylating S648. Our results demonstrate a diversity of NHE1:CaM interaction modes and suggest that CaM may contribute to NHE1 dimerization and thereby augment NHE1 regulation. We propose that a similar structural diversity is of relevance to many other CaM complexes., Competing Interests: LS, AP, EP, MS, KK, JO, BK, SP No competing interests declared, (© 2021, Sjøgaard-Frich et al.)- Published
- 2021
- Full Text
- View/download PDF
37. Putting Warburg to work: how imaging of tumour acidosis could help predict metastatic potential in breast cancer.
- Author
-
Rolver MG and Pedersen SF
- Subjects
- Humans, Hydrogen-Ion Concentration, Magnetic Resonance Imaging, Acidosis, Breast Neoplasms diagnostic imaging
- Abstract
Solid tumours are often highly acidic compared to normal tissue, and tumour extracellular acidosis contributes to multiple aspects of cancer progression. Now, Anemone et al. in this issue of the British Journal of Cancer provide in vivo evidence that the degree to which various breast cancer cell lines acidify their environment correlates with their ability to metastasise to the lungs. This indicates that measurements of tumour extracellular acidosis have the potential to become a clinical tool for assessing the risk of metastasis.
- Published
- 2021
- Full Text
- View/download PDF
38. The intracellular lipid-binding domain of human Na + /H + exchanger 1 forms a lipid-protein co-structure essential for activity.
- Author
-
Hendus-Altenburger R, Vogensen J, Pedersen ES, Luchini A, Araya-Secchi R, Bendsoe AH, Prasad NS, Prestel A, Cardenas M, Pedraz-Cuesta E, Arleth L, Pedersen SF, and Kragelund BB
- Subjects
- Animals, CHO Cells, Circular Dichroism, Cricetinae, Cricetulus, Humans, Protein Binding, Protein Conformation, Protein Domains, Sodium-Hydrogen Exchanger 1 metabolism, Lipids chemistry, Sodium-Hydrogen Exchanger 1 chemistry
- Abstract
Dynamic interactions of proteins with lipid membranes are essential regulatory events in biology, but remain rudimentarily understood and particularly overlooked in membrane proteins. The ubiquitously expressed membrane protein Na
+ /H+ -exchanger 1 (NHE1) regulates intracellular pH (pHi ) with dysregulation linked to e.g. cancer and cardiovascular diseases. NHE1 has a long, regulatory cytosolic domain carrying a membrane-proximal region described as a lipid-interacting domain (LID), yet, the LID structure and underlying molecular mechanisms are unknown. Here we decompose these, combining structural and biophysical methods, molecular dynamics simulations, cellular biotinylation- and immunofluorescence analysis and exchanger activity assays. We find that the NHE1-LID is intrinsically disordered and, in presence of membrane mimetics, forms a helical αα-hairpin co-structure with the membrane, anchoring the regulatory domain vis-a-vis the transport domain. This co-structure is fundamental for NHE1 activity, as its disintegration reduced steady-state pHi and the rate of pHi recovery after acid loading. We propose that regulatory lipid-protein co-structures may play equally important roles in other membrane proteins.- Published
- 2020
- Full Text
- View/download PDF
39. Cancer Cell Acid Adaptation Gene Expression Response Is Correlated to Tumor-Specific Tissue Expression Profiles and Patient Survival.
- Author
-
Yao J, Czaplinska D, Ialchina R, Schnipper J, Liu B, Sandelin A, and Pedersen SF
- Abstract
The acidic pH of the tumor microenvironment plays a critical role in driving cancer development toward a more aggressive phenotype, but the underlying mechanisms are unclear. To this end, phenotypic and genotypic changes induced by adaptation of cancer cells to chronic acidosis have been studied. However, the generality of acid adaptation patterns across cell models and their correlation to the molecular phenotypes and aggressiveness of human cancers are essentially unknown. Here, we define an acid adaptation expression response shared across three cancer cell models, dominated by metabolic rewiring, extracellular matrix remodeling, and altered cell cycle regulation and DNA damage response. We find that many genes which are upregulated by acid adaptation are significantly correlated to patient survival, and more generally, that there are clear correlations between acid adaptation expression response and gene expression change between normal and tumor tissues, for a large subset of cancer patients. Our data support the notion that tumor microenvironment acidity is one of the key factors driving the selection of aggressive cancer cells in human patient tumors, yet it also induces a growth-limiting genotype that likely limits cancer cell growth until the cells are released from acidosis, for instance during invasion.
- Published
- 2020
- Full Text
- View/download PDF
40. The COVID-19 pandemic from an A&E perspective.
- Author
-
Wammer ACP, Pedersen SF, Hatleskog ML, and Hegna BW
- Subjects
- Betacoronavirus, COVID-19, Humans, SARS-CoV-2, Coronavirus Infections, Pandemics, Pneumonia, Viral
- Published
- 2020
- Full Text
- View/download PDF
41. O-glycan initiation directs distinct biological pathways and controls epithelial differentiation.
- Author
-
Bagdonaite I, Pallesen EM, Ye Z, Vakhrushev SY, Marinova IN, Nielsen MI, Kramer SH, Pedersen SF, Joshi HJ, Bennett EP, Dabelsteen S, and Wandall HH
- Subjects
- Cell Differentiation, Epithelium metabolism, Glycosylation, Humans, Polysaccharides, Protein Processing, Post-Translational, N-Acetylgalactosaminyltransferases genetics, N-Acetylgalactosaminyltransferases metabolism
- Abstract
Post-translational modifications (PTMs) greatly expand the function and potential for regulation of protein activity, and O-glycosylation is among the most abundant and diverse PTMs. Initiation of O-GalNAc glycosylation is regulated by 20 distinct GalNAc-transferases (GalNAc-Ts), and deficiencies in individual GalNAc-Ts are associated with human disease, causing subtle but distinct phenotypes in model organisms. Here, we generate a set of isogenic keratinocyte cell lines lacking either of the three dominant and differentially expressed GalNAc-Ts. Through the ability of keratinocytes to form epithelia, we investigate the phenotypic consequences of the loss of individual GalNAc-Ts. Moreover, we probe the cellular responses through global transcriptomic, differential glycoproteomic, and differential phosphoproteomic analyses. We demonstrate that loss of individual GalNAc-T isoforms causes distinct epithelial phenotypes through their effect on specific biological pathways; GalNAc-T1 targets are associated with components of the endomembrane system, GalNAc-T2 targets with cell-ECM adhesion, and GalNAc-T3 targets with epithelial differentiation. Thus, GalNAc-T isoforms serve specific roles during human epithelial tissue formation., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
42. Avidity within the N-terminal anchor drives α-synuclein membrane interaction and insertion.
- Author
-
Cholak E, Bugge K, Khondker A, Gauger K, Pedraz-Cuesta E, Pedersen ME, Bucciarelli S, Vestergaard B, Pedersen SF, Rheinstädter MC, Langkilde AE, and Kragelund BB
- Subjects
- Animals, Cells, Cultured, Humans, Mammals metabolism, Membrane Fusion physiology, Cell Membrane metabolism, alpha-Synuclein metabolism
- Abstract
In the brain, α-synuclein (aSN) partitions between free unbound cytosolic and membrane bound forms modulating both its physiological and pathological role and complicating its study due to structural heterogeneity. Here, we use an interdisciplinary, synergistic approach to characterize the properties of aSN:lipid mixtures, isolated aSN:lipid co-structures, and aSN in mammalian cells. Enabled by the isolation of the membrane-bound state, we show that within the previously described N-terminal membrane anchor, membrane interaction relies both on an N-terminal tail (NTT) head group layer insertion of 14 residues and a folded-upon-binding helix at the membrane surface. Both binding events must be present; if, for example, the NTT insertion is lost, the membrane affinity of aSN is severely compromised and formation of aSN:lipid co-structures hampered. In mammalian cells, compromised cooperativity results in lowered membrane association. Thus, avidity within the N-terminal anchor couples N-terminal insertion and helical surface binding, which is crucial for aSN membrane interaction and cellular localization, and may affect membrane fusion., (© 2020 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)
- Published
- 2020
- Full Text
- View/download PDF
43. Yeast recombinant production of intact human membrane proteins with long intrinsically disordered intracellular regions for structural studies.
- Author
-
Kassem N, Kassem MM, Pedersen SF, Pedersen PA, and Kragelund BB
- Subjects
- Humans, Membrane Proteins biosynthesis, Protein Conformation, Receptors, Somatotropin chemistry, Recombinant Proteins biosynthesis, Sodium-Hydrogen Exchangers chemistry, Yeasts genetics, Intrinsically Disordered Proteins chemistry, Membrane Proteins chemistry, Recombinant Proteins chemistry, Saccharomyces cerevisiae genetics
- Abstract
Membrane proteins exist in lipid bilayers and mediate solute transport, signal transduction, cell-cell communication and energy conversion. Their activities are fundamental for life, which make them prominent subjects of study, but access to only a limited number of high-resolution structures complicates their mechanistic understanding. The absence of such structures relates mainly to difficulties in expressing and purifying high quality membrane protein samples in large quantities. An additional layer of complexity stems from the presence of intra- and/or extra-cellular domains constituted by unstructured intrinsically disordered regions (IDR), which can be hundreds of residues long. Although IDRs form key interaction hubs that facilitate biological processes, these are regularly removed to enable structural studies. To advance mechanistic insight into intact intrinsically disordered membrane proteins, we have developed a protocol for their purification. Using engineered yeast cells for optimized expression and purification, we have purified to homogeneity two very different human membrane proteins each with >300 residues long IDRs; the sodium proton exchanger 1 and the growth hormone receptor. Subsequent to their purification we have further explored their incorporation into membrane scaffolding protein nanodiscs, which will enable future structural studies., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
44. TGFβ Signaling Increases Net Acid Extrusion, Proliferation and Invasion in Panc-1 Pancreatic Cancer Cells: SMAD4 Dependence and Link to Merlin/NF2 Signaling.
- Author
-
Malinda RR, Zeeberg K, Sharku PC, Ludwig MQ, Pedersen LB, Christensen ST, and Pedersen SF
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer-related death, with a 5-year survival of <10% and severely limited treatment options. PDAC hallmarks include profound metabolic acid production and aggressive local proliferation and invasiveness. This phenotype is supported by upregulated net acid extrusion and epithelial-to-mesenchymal transition (EMT), the latter typically induced by aberrant transforming growth factor-β (TGFβ) signaling. It is, however, unknown whether TGFβ-induced EMT and upregulation of acid extrusion are causally related. Here, we show that mRNA and protein expression of the net acid extruding transporters Na
+ /H+ exchanger 1 (NHE1, SLC9A1) and Na+ , HCO 3 - cotransporter 1 (NBCn1, SLC4A7) are increased in a panel of human PDAC cell lines compared to immortalized human pancreatic ductal epithelial (HPDE) cells. Treatment of Panc-1 cells (which express SMAD4, required for canonical TGFβ signaling) with TGFβ-1 for 48 h elicited classical EMT with down- and upregulation of epithelial and mesenchymal markers, respectively, in a manner inhibited by SMAD4 knockdown. Accordingly, less pronounced EMT was induced in BxPC-3 cells, which do not express SMAD4. TGFβ-1 treatment elicited a SMAD4-dependent increase in NHE1 expression, and a smaller, SMAD4-independent increase in NBCn1 in Panc-1 cells. Consistent with this, TGFβ-1 treatment led to elevated intracellular pH and increased net acid extrusion capacity in Panc-1 cells, but not in BxPC-3 cells, in an NHE1-dependent manner. Proliferation was increased in Panc-1 cells and decreased in BxPC-3 cells, upon TGFβ-1 treatment, and this, as well as EMT per se , was unaffected by NHE1- or NBCn1 inhibition. TGFβ-1-induced EMT was associated with a 4-fold increase in Panc-1 cell invasiveness, which further increased ~10-fold upon knockdown of the tumor suppressor Merlin (Neurofibromatosis type 2). Knockdown of NHE1 or NBCn1 abolished Merlin-induced invasiveness, but not that induced by TGFβ-1 alone. In conclusion, NHE1 and NBCn1 expression and NHE-dependent acid extrusion are upregulated during TGFβ-1-induced EMT of Panc-1 cells. NHE1 upregulation is SMAD4-dependent, and SMAD4-deficient BxPC-3 cells show no change in pHi regulation. NHE1 and NBCn1 are not required for EMT per se or EMT-associated proliferation changes, but are essential for the potentiation of invasiveness induced by Merlin knockdown., (Copyright © 2020 Malinda, Zeeberg, Sharku, Ludwig, Pedersen, Christensen and Pedersen.)- Published
- 2020
- Full Text
- View/download PDF
45. SARS-CoV-2: a storm is raging.
- Author
-
Pedersen SF and Ho YC
- Subjects
- Age Factors, COVID-19, Coronavirus Infections complications, Coronavirus Infections physiopathology, Coronavirus Infections virology, Cytokines blood, Disease Progression, Humans, Lymphocytes cytology, Lymphocytes immunology, Pandemics, Pneumonia, Viral complications, Pneumonia, Viral physiopathology, Pneumonia, Viral virology, Respiratory Distress Syndrome etiology, SARS-CoV-2, Severity of Illness Index, Betacoronavirus immunology, Coronavirus Infections immunology, Cytokines immunology, Pneumonia, Viral immunology
- Abstract
The pandemic coronavirus infectious disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is rapidly spreading across the globe. In this issue of the JCI, Chen and colleagues compared the clinical and immunological characteristics between moderate and severe COVID-19. The authors found that respiratory distress on admission is associated with unfavorable outcomes. Increased cytokine levels (IL-6, IL-10, and TNF-α), lymphopenia (in CD4+ and CD8+ T cells), and decreased IFN-γ expression in CD4+ T cells are associated with severe COVID-19. Overall, this study characterized the cytokine storm in severe COVID-19 and provides insights into immune therapeutics and vaccine design.
- Published
- 2020
- Full Text
- View/download PDF
46. Why Warburg Works: Lactate Controls Immune Evasion through GPR81.
- Author
-
Lundø K, Trauelsen M, Pedersen SF, and Schwartz TW
- Subjects
- Antigen-Presenting Cells, Humans, Immune Evasion, Lactic Acid, Receptors, G-Protein-Coupled, Breast Neoplasms, Tumor Microenvironment
- 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., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
47. Pyrazine ring-based Na + /H + exchanger (NHE) inhibitors potently inhibit cancer cell growth in 3D culture, independent of NHE1.
- Author
-
Rolver MG, Elingaard-Larsen LO, Andersen AP, Counillon L, and Pedersen SF
- Subjects
- Apoptosis, Autophagy, Cell Proliferation, Endoplasmic Reticulum Stress, Humans, MCF-7 Cells, Neoplasms metabolism, Sodium-Hydrogen Exchanger 1 genetics, Sodium-Hydrogen Exchanger 1 metabolism, Spheroids, Cellular metabolism, Amiloride pharmacology, Antineoplastic Agents pharmacology, Guanidines pharmacology, Spheroids, Cellular drug effects, Sulfones pharmacology
- Abstract
The Na
+ /H+ exchanger-1 (NHE1) supports tumour growth, making NHE1 inhibitors of interest in anticancer therapy, yet their molecular effects are incompletely characterized. Here, we demonstrate that widely used pyrazinoylguanidine-type NHE1 inhibitors potently inhibit growth and survival of cancer cell spheroids, in a manner unrelated to NHE1 inhibition. Cancer and non-cancer cells were grown as 3-dimensional (3D) spheroids and treated with pyrazinoylguanidine-type (amiloride, 5-(N-ethyl-N-isopropyl)-amiloride (EIPA), 5-(N,N-dimethyl)-amiloride (DMA), and 5-(N,N-hexamethylene)-amiloride (HMA)) or benzoylguanidine-type (eniporide, cariporide) NHE1 inhibitors for 2-7 days, followed by analyses of viability, compound accumulation, and stress- and death-associated signalling. EIPA, DMA and HMA dose-dependently reduced breast cancer spheroid viability while cariporide and eniporide had no effect. Although both compound types inhibited NHE1, the toxic effects were NHE1-independent, as inhibitor-induced viability loss was unaffected by NHE1 CRISPR/Cas9 knockout. EIPA and HMA accumulated extensively in spheroids, and this was associated with marked vacuolization, apparent autophagic arrest, ER stress, mitochondrial- and DNA damage and poly-ADP-ribose-polymerase (PARP) cleavage, indicative of severe stress and paraptosis-like cell death. Pyrazinoylguanidine-induced cell death was partially additive to that induced by conventional anticancer therapies and strongly additive to extracellular-signal-regulated-kinase (ERK) pathway inhibition. Thus, in addition to inhibiting NHE1, pyrazinoylguanidines exert potent, NHE1-independent cancer cell death, pointing to a novel relevance for these compounds in anticancer therapy.- Published
- 2020
- Full Text
- View/download PDF
48. The Vacuolar H + ATPase α3 Subunit Negatively Regulates Migration and Invasion of Human Pancreatic Ductal Adenocarcinoma Cells.
- Author
-
Flinck M, Hagelund S, Gorbatenko A, Severin M, Pedraz-Cuesta E, Novak I, Stock C, and Pedersen SF
- Subjects
- Autophagy drug effects, Autophagy genetics, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Gene Knockdown Techniques, Humans, Hydrogen-Ion Concentration, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Neoplasm Invasiveness genetics, Pancreatic Neoplasms pathology, Protease Inhibitors pharmacology, Protein Subunits genetics, Signal Transduction drug effects, Signal Transduction genetics, Up-Regulation, Vacuolar Proton-Translocating ATPases antagonists & inhibitors, Carcinoma, Pancreatic Ductal metabolism, Cell Movement genetics, Pancreatic Neoplasms metabolism, Protein Subunits metabolism, Vacuolar Proton-Translocating ATPases metabolism
- Abstract
Increased metabolic acid production and upregulation of net acid extrusion render pH homeostasis profoundly dysregulated in many cancers. Plasma membrane activity of vacuolar H
+ ATPases (V-ATPases) has been implicated in acid extrusion and invasiveness of some cancers, yet often on the basis of unspecific inhibitors. Serving as a membrane anchor directing V-ATPase localization, the a subunit of the V0 domain of the V-ATPase (ATP6V0a1-4) is particularly interesting in this regard. Here, we map the regulation and roles of ATP6V0a3 in migration, invasion, and growth in pancreatic ductal adenocarcinoma (PDAC) cells. a3 mRNA and protein levels were upregulated in PDAC cell lines compared to non-cancer pancreatic epithelial cells. Under control conditions, a3 localization was mainly endo-/lysosomal, and its knockdown had no detectable effect on pHi regulation after acid loading. V-ATPase inhibition, but not a3 knockdown, increased HIF-1 expression and decreased proliferation and autophagic flux under both starved and non-starved conditions, and spheroid growth of PDAC cells was also unaffected by a3 knockdown. Strikingly, a3 knockdown increased migration and transwell invasion of Panc-1 and BxPC-3 PDAC cells, and increased gelatin degradation in BxPC-3 cells yet decreased it in Panc-1 cells. We conclude that in these PDAC cells, a3 is upregulated and negatively regulates migration and invasion, likely in part via effects on extracellular matrix degradation., Competing Interests: The authors declare no conflict of interest.- Published
- 2020
- Full Text
- View/download PDF
49. The γ-hydroxybutyric acid (GHB) analogue NCS-382 is a substrate for both monocarboxylate transporters subtypes 1 and 4.
- Author
-
Thiesen L, Belew ZM, Griem-Krey N, Pedersen SF, Crocoll C, Nour-Eldin HH, and Wellendorph P
- Subjects
- Animals, Cell Line, Humans, Hydroxybutyrates, Ligands, Monocarboxylic Acid Transporters genetics, Muscle Proteins genetics, Oocytes metabolism, Symporters genetics, Xenopus laevis, Benzocycloheptenes pharmacology, Monocarboxylic Acid Transporters metabolism, Muscle Proteins metabolism, Symporters metabolism
- Abstract
The small-molecule ligand (E)-2-(5-hydroxy-5,7,8,9-tetrahydro-6H-benzo[7]annulen-6-ylidene)acetic acid (NCS-382) is an analogue of γ-hydroxybutyric acid (GHB) and is widely used for probing the brain-specific GHB high-affinity binding sites. To reach these, brain uptake is imperative, and it is therefore important to understand the molecular mechanisms of NCS-382 transport in order to direct in vivo studies. In this study, we hypothesized that NCS-382 is a substrate for the monocarboxylate transporter subtype 1 (MCT1) which is known to mediate blood-brain barrier (BBB) permeation of GHB. For this purpose, we investigated NCS-382 uptake by MCT subtypes endogenously expressed in tsA201 and MDA-MB-231 cell lines in assays of radioligand-based competition and fluorescence-based intracellular pH measurements. To further verify the results, we measured NCS-382 uptake by means of mass spectrometry in Xenopus laevis oocytes heterologously expressing MCT subtypes. As expected, we found that NCS-382 is a substrate for MCT1 with half-maximal effective concentrations in the low millimolar range. Surprisingly, NCS-382 also showed substrate activity at MCT4 as well as uptake in water-injected oocytes, suggesting a component of passive diffusion. In conclusion, transport of NCS-382 across membranes differs from GHB as it also involves MCT4 and/or passive diffusion. This should be taken into consideration when designing pharmacological studies with this compound and its closely related analogues. The combination of MCT assays used here exemplifies a setup that may be suitable for a reliable characterization of MCT ligands in general., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
50. The Acidic Tumor Microenvironment as a Driver of Cancer.
- Author
-
Boedtkjer E and Pedersen SF
- Subjects
- Acids metabolism, Animals, Humans, Hydrogen-Ion Concentration, Neoplasm Metastasis, Neoplasms pathology, Signal Transduction, Neoplasms metabolism, Tumor Microenvironment
- Abstract
Acidic metabolic waste products accumulate in the tumor microenvironment because of high metabolic activity and insufficient perfusion. In tumors, the acidity of the interstitial space and the relatively well-maintained intracellular pH influence cancer and stromal cell function, their mutual interplay, and their interactions with the extracellular matrix. Tumor pH is spatially and temporally heterogeneous, and the fitness advantage of cancer cells adapted to extracellular acidity is likely particularly evident when they encounter less acidic tumor regions, for instance, during invasion. Through complex effects on genetic stability, epigenetics, cellular metabolism, proliferation, and survival, the compartmentalized pH microenvironment favors cancer development. Cellular selection exacerbates the malignant phenotype, which is further enhanced by acid-induced cell motility, extracellular matrix degradation, attenuated immune responses, and modified cellular and intercellular signaling. In this review, we discuss how the acidity of the tumor microenvironment influences each stage in cancer development, from dysplasia to full-blown metastatic disease.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.