1
|
Benatzy Y, Palmer MA, Lütjohann D, Ohno RI, Kampschulte N, Schebb NH, Fuhrmann DC, Snodgrass RG, Brüne B. ALOX15B controls macrophage cholesterol homeostasis via lipid peroxidation, ERK1/2 and SREBP2. Redox Biol 2024; 72:103149. [PMID: 38581859 PMCID: PMC11002893 DOI: 10.1016/j.redox.2024.103149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
Macrophage cholesterol homeostasis is crucial for health and disease and has been linked to the lipid-peroxidizing enzyme arachidonate 15-lipoxygenase type B (ALOX15B), albeit molecular mechanisms remain obscure. We performed global transcriptome and immunofluorescence analysis in ALOX15B-silenced primary human macrophages and observed a reduction of nuclear sterol regulatory element-binding protein (SREBP) 2, the master transcription factor of cellular cholesterol biosynthesis. Consequently, SREBP2-target gene expression was reduced as were the sterol biosynthetic intermediates desmosterol and lathosterol as well as 25- and 27-hydroxycholesterol. Mechanistically, suppression of ALOX15B reduced lipid peroxidation in primary human macrophages and thereby attenuated activation of mitogen-activated protein kinase ERK1/2, which lowered SREBP2 abundance and activity. Low nuclear SREBP2 rendered both, ALOX15B-silenced and ERK1/2-inhibited macrophages refractory to SREBP2 activation upon blocking the NPC intracellular cholesterol transporter 1. These studies suggest a regulatory mechanism controlling macrophage cholesterol homeostasis based on ALOX15B-mediated lipid peroxidation and concomitant ERK1/2 activation.
Collapse
Affiliation(s)
- Yvonne Benatzy
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Megan A Palmer
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Rei-Ichi Ohno
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nadja Kampschulte
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Dominik C Fuhrmann
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany.
| | - Ryan G Snodgrass
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany; Western Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Davis, CA, USA.
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany.
| |
Collapse
|
2
|
van Brakel L, Mensink RP, Lütjohann D, Plat J. Plant stanol consumption increases anti-COVID-19 antibody responses, independent of changes in serum cholesterol concentrations: a randomized controlled trial. Am J Clin Nutr 2024; 119:969-980. [PMID: 38278364 DOI: 10.1016/j.ajcnut.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND People with overweight/obesity generally have impaired immune responses, resulting among others in increased risk of severe complaints and hospitalization after infections with severe acute respiratory syndrome coronavirus 2 (COVID-19), as well as decreased antibody production after vaccinations. Plant stanol ester previously increased the combined IgM/IgG antibody titers toward a hepatitis A vaccination in patients with allergic asthma, but the underlying mechanism is unknown. OBJECTIVES We evaluated whether plant stanol ester consumption improved the immune response in subjects with overweight/obesity after a COVID-19 vaccination. METHODS A double-blind, randomized, placebo-controlled trial was performed. Thirty-two subjects with overweight/obesity consumed products with added plant stanols (4 g/d; provided as plant stanol ester) or control ≥2 wk before receiving their COVID-19 vaccination until 4 wk after vaccination. Antibody titers were analyzed weekly and statistically analyzed using mixed models. Serum metabolic markers and cytokine profiles were also analyzed. RESULTS IgM concentrations against the COVID-19 Spike protein were increased in the plant stanol ester group compared with the control group, with the largest difference observed 2 wk after vaccination [31.2 (0.43, 62.1) BAU/mL, or +139%; Group × Time: P = 0.031]. Subjects that produced very low IgM antibodies produced, as expected, hardly any IgG antibodies. In those with IgG seroconversion, IgG Spike concentrations were also increased in the plant stanol ester group compared with the control group [71.3 (2.51, 140.1) BAU/mL; Group P = 0.043]. Stimulated cytokine concentrations decreased in the plant stanol ester group compared with the control group in all 3 cytokine domains (that is, proinflammatory, T helper [Th1]/Th17, and Th2/regulatory T cells). Between-group differences in serum LDL cholesterol or other metabolic markers were not observed. CONCLUSIONS Consuming plant stanols (4 g/d) affects immune responses to COVID-19 vaccinations, translating into increased serum anti-COVID-19 IgM concentrations in subjects with overweight/obesity. Only in IgG seroconverted subjects, serum anti-COVID-19 IgG concentrations also increase. These effects are independent of reductions in LDL cholesterol. These results suggest that this high-risk group for COVID-19 complications could benefit from plant stanol consumption. This trial was registered at clinicaltrials.gov as NCT04844346.
Collapse
Affiliation(s)
- Lieve van Brakel
- Department of Nutrition and Movement Sciences, NUTRIM School of Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.
| | - Ronald P Mensink
- Department of Nutrition and Movement Sciences, NUTRIM School of Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, NUTRIM School of Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Spinedi M, Clark C, Zullo L, Kerksiek A, Pistis G, Castelao E, von Gunten A, Preisig M, Lütjohann D, Popp J. Cholesterol-metabolism, plant sterols, and long-term cognitive decline in older people - Effects of sex and APOEe4. iScience 2024; 27:109013. [PMID: 38327787 PMCID: PMC10847741 DOI: 10.1016/j.isci.2024.109013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
Neurodegenerative, vascular, and dementia diseases are linked to dysregulations in cholesterol metabolism. Dietary plant sterols, or phytosterols, may interfere to neurodegeneration and cognitive decline, and have cholesterol-lowering, anti-inflammatory, and antioxidant qualities. Here, we investigated the potential associations between circulating cholesterol precursors and metabolites, triglycerides, and phytosterols with cognitive decline in older people by performing multivariate analysis on 246 participants engaged in a population-based prospective study. In our analysis we considered the potential effect of sex and APOEe4. We reveal particular dysregulations of diet-derived phytosterols and endogenous cholesterol synthesis and metabolism, and their variations over time linked to cognitive decline in the general population. These results are significant to the development of interventions to avoid cognitive decline in older adults and suggest that levels of circulating sterols should be taken into account when evaluating risk.
Collapse
Affiliation(s)
- Matteo Spinedi
- University Hospital of Psychiatry and University of Zürich, Zürich, Switzerland
| | - Christopher Clark
- University Hospital of Psychiatry and University of Zürich, Zürich, Switzerland
| | - Leonardo Zullo
- Service of Old Age Psychiatry, Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Giorgio Pistis
- Psychiatric Epidemiology and Psychopathology Research Center, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Enrique Castelao
- Psychiatric Epidemiology and Psychopathology Research Center, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Armin von Gunten
- Service of Old Age Psychiatry, Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Martin Preisig
- Psychiatric Epidemiology and Psychopathology Research Center, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Julius Popp
- University Hospital of Psychiatry and University of Zürich, Zürich, Switzerland
- Service of Old Age Psychiatry, Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Schumacher S, Klose L, Lambertz J, Lütjohann D, Biemann R, Kuerten S, Fester L. The mitochondrial protease PARL is required for spermatogenesis. Commun Biol 2024; 7:44. [PMID: 38182793 PMCID: PMC10770312 DOI: 10.1038/s42003-023-05703-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/13/2023] [Indexed: 01/07/2024] Open
Abstract
Mitochondrial function plays an important role in the maintenance of male fertility. However, the mechanisms underlying mitochondrial defect-related infertility remain mostly unclear. Here we show that a deficiency of PARL (Parl-/-), a mitochondrial protease, causes complete arrest of spermatogenesis during meiosis I. PARL deficiency led to severe downregulation of proteins of respiratory chain complex IV in testes that did not occur in other tested organs, causing a deficit in complex IV activity and ATP production. Furthermore, Parl-/- testes showed an almost complete loss of HSD17B3, a protein of the sER responsible for the last step in testosterone synthesis. While testosterone production appeared to be restored by overexpression of HSD17B12, loss of the canonical testosterone synthesis led to an upregulation of luteinizing hormone (LH) and of LH-regulated responses. These results suggest an important impact of the downstream regulation of mitochondrial defects that manifest in a cell-type-specific manner and extend beyond mitochondria.
Collapse
Affiliation(s)
- Sarah Schumacher
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115, Bonn, Germany.
| | - Laura Klose
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115, Bonn, Germany
| | - Jessica Lambertz
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115, Bonn, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, 04103, Leipzig, Germany
| | - Stefanie Kuerten
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115, Bonn, Germany
| | - Lars Fester
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
5
|
Deng X, Lin B, Wang F, Xu P, Wang N. Specnuezhenide Ameliorates Age-Related Hepatic Lipid Accumulation via Modulating Bile Acid Homeostasis and Gut Microbiota in D-Galactose-Induced Mice. Metabolites 2023; 13:960. [PMID: 37623903 PMCID: PMC10456809 DOI: 10.3390/metabo13080960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Age-related hepatic lipid accumulation has become a major health problem in the elderly population. Specnuezhenide (SPN) is a major active iridoid glycoside from an edible herb Fructus Ligustri Lucidi, which is commonly used for preventing age-related diseases. However, the beneficial effects of SPN on age-related liver injury remain unknown. This study aimed to reveal the effect of SPN on age-related hepatic lipid accumulation and the underlying mechanism. D-galactose (D-gal)-induced aging mice were treated with vehicle or SPN for 12 weeks. Treatment of SPN decreased lipid accumulation and inflammation in the liver of D-gal-induced mice. Untargeted and targeted metabolomics showed that the SPN could regulate the bile acid (BA) synthesis pathway and restore the BA compositions in serum, livers, and feces of the D-gal-induced mice. Furthermore, SPN enhanced the protein and mRNA levels of hepatic BAs synthesis enzymes cytochrome P45027A1, cytochrome P4507A1, cytochrome P4507B1, and cytochrome P4508B1. Meanwhile, SPN alleviated D-gal-induced gut dysbiosis and reversed the proportions of microbes associated with bile salt hydrolase activity, including Lactobacillus, Ruminiclostridium, and Butyrivibrio. Our study revealed that SPN attenuated age-related hepatic lipid accumulation by improving BA profiles via modulating hepatic BA synthesis enzymes and gut microbiota.
Collapse
Affiliation(s)
- Xuehui Deng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310007, China; (X.D.)
| | - Bingfeng Lin
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China; (B.L.)
| | - Fang Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310007, China; (X.D.)
| | - Pingcui Xu
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China; (B.L.)
| | - Nani Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310007, China; (X.D.)
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China; (B.L.)
| |
Collapse
|
6
|
Šošić-Jurjević B, Lütjohann D, Trifunović S, Pavlović S, Borković Mitić S, Jovanović L, Ristić N, Marina L, Ajdžanović V, Filipović B. Differences in Cholesterol Metabolism, Hepato-Intestinal Aging, and Hepatic Endocrine Milieu in Rats as Affected by the Sex and Age. Int J Mol Sci 2023; 24:12624. [PMID: 37628805 PMCID: PMC10454938 DOI: 10.3390/ijms241612624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Age and sex influence serum cholesterol levels, but the underlying mechanisms remain unclear. To investigate further, we measured cholesterol, precursors (surrogate synthesis markers), degradation products (oxysterols and bile acid precursors) in serum, the liver, jejunum, and ileum, as well as serum plant sterols (intestinal absorption markers) in male and female Wistar rats (4 and 24 months old). The analysis of histomorphometric and oxidative stress parameters (superoxide dismutase, catalase, glutathione-related enzyme activities, lipid peroxide, and protein carbonyl concentrations) in the liver and jejunum offered further insights into the age- and sex-related differences. The hepatic gene expression analysis included AR, ERα, and sex-specific growth hormone-regulated (Cyp2c11 and Cyp2c12) and thyroid-responsive (Dio1, Tbg, and Spot 14) genes by qPCR. We observed age-related changes in both sexes, with greater prominence in females. Aged females had significantly higher serum cholesterol (p < 0.05), jejunum cholesterol (p < 0.05), and serum plant sterols (p < 0.05). They exhibited poorer hepato-intestinal health compared with males, which was characterized by mild liver dysfunction (hydropic degeneration, increased serum ALT, p < 0.05, and decreased activity of some antioxidant defense enzymes, p < 0.05), mononuclear inflammation in the jejunal lamina propria, and age-related decreases in jejunal catalase and glutathione peroxidase activity (p < 0.05). Aged females showed increased levels of 27-hydroxycholesterol (p < 0.05) and upregulated ERα gene expression (p < 0.05) in the liver. Our study suggests that the more significant age-related increase in serum cholesterol in females is associated with poorer hepato-intestinal health and increased jejunal cholesterol absorption. The local increase in 27-hydroxycholesterol during aging might reduce the hepatoprotective effects of endogenous estrogen in the female liver.
Collapse
Affiliation(s)
- Branka Šošić-Jurjević
- Department of Cytology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (S.T.); (N.R.); (V.A.); (B.F.)
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany;
| | - Svetlana Trifunović
- Department of Cytology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (S.T.); (N.R.); (V.A.); (B.F.)
| | - Slađan Pavlović
- Department of Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (S.P.); (S.B.M.)
| | - Slavica Borković Mitić
- Department of Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (S.P.); (S.B.M.)
| | - Ljubiša Jovanović
- Department of Pathology and Medical Cytology, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Dr. Koste Todorovića 26, 11000 Belgrade, Serbia;
| | - Nataša Ristić
- Department of Cytology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (S.T.); (N.R.); (V.A.); (B.F.)
| | - Ljiljana Marina
- National Centre for Infertility and Endocrinology of Gender, Clinic for Endocrinology, Diabetes and Metabolic Diseases, Faculty of Medicine, University of Belgrade, Koste Todorovića 6, 11000 Belgrade, Serbia;
| | - Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (S.T.); (N.R.); (V.A.); (B.F.)
| | - Branko Filipović
- Department of Cytology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (S.T.); (N.R.); (V.A.); (B.F.)
| |
Collapse
|
7
|
Peripubertal soy isoflavone consumption leads to subclinical hypothyroidism in male Wistar rats. J Dev Orig Health Dis 2023; 14:209-222. [PMID: 36017706 DOI: 10.1017/s2040174422000496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Exposure to endocrine-disrupting chemicals during critical windows of development may lead to functional abnormalities in adulthood. Isoflavones are a flavonoid group of phytoestrogens that are recognized by their estrogenic activity and are highly abundant in soybean. Since the thyroid gland presents estrogen receptors and infants, toddlers and teenagers may consume isoflavones from soy-based infant formula and beverages as alternatives to animal milk, we propose to investigate the potential effects of relevant concentrations of soy isoflavones in the regulation of the hypothalamic-pituitary (HP) thyroid axis using peripubertal male rats as an experimental model. Thirty-two 23-day-old male rats were exposed to 0.5, 5, or 50 mg of soy isoflavones/kg from weaning to 60 days of age, when they were euthanized, and the tissues were collected to evaluate the mRNA expression of genes involved in the regulation of the HP thyroid axis and dosages of thyroid hormones (THs). Serum TSH concentrations were increased, while alterations were not observed in serum concentrations of triiodothyronine and thyroxine. Regarding mRNA gene expression, Mct-8 was increased in the hypothalamus, Mct-8, Thra1, and Thrb2 were decreased in the pituitary, and Nis and Pds were reduced in the thyroid. In the heart, Mct8 and Thrb2 were increased, and Thra1 was decreased. In the liver, Mct8, Thra1, and Thrb2 were decreased. These results suggest that the consumption of relevant doses of soy isoflavones during the peripubertal period in males may induce subclinical hypothyroidism, with alterations in the regulation of the HP thyroid axis, modulation of TH synthesis, and peripheral alterations in TH target organs.
Collapse
|
8
|
A Transient Inflammatory Response Induced by Lipopolysaccharide Infusion Lowers Markers of Endogenous Cholesterol and Bile Acid Synthesis in Healthy Normocholesterolemic Young Men. Biomedicines 2023; 11:biomedicines11010126. [PMID: 36672634 PMCID: PMC9855383 DOI: 10.3390/biomedicines11010126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/27/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Inflammation is associated with changes in plasma lipids, lipoproteins, and cholesterol efflux capacity (CEC). It is unknown if the changes in lipids and lipoproteins during inflammation are related to changes in cholesterol absorption, synthesis, and bile acid synthesis. We, therefore, examined the effects of acute lipopolysaccharide (LPS)-induced transient systemic inflammation on lipids, lipoproteins, CEC, and markers of cholesterol metabolism. We also evaluated whether markers for cholesterol metabolism at baseline predict the intensity of the inflammatory response. Eight healthy young subjects received LPS infusion, and blood was sampled for the following 24 h. In addition to lipids, lipoproteins, and CEC, we also measured markers for cholesterol absorption and synthesis, bile acid synthesis, and inflammation. Compared with baseline, plasma total cholesterol, low-density lipoprotein cholesterol, and CEC decreased, while triglycerides increased in the 24 h following LPS infusion. TC-standardized levels of cholesterol synthesis markers (lathosterol, lanosterol, and desmosterol) and a bile acid synthesis marker (7α-OH-cholesterol) also decreased, with no changes in cholesterol absorption markers (campesterol, sitosterol, and cholestanol). Baseline TC-standardized levels of desmosterol and 7α-OH-cholesterol were positively correlated with concentrations of various inflammatory markers. Changes in TC-standardized desmosterol and 7α-OH-cholesterol were negatively correlated with concentrations of inflammatory markers. LPS infusion reduced endogenous cholesterol synthesis and bile acid synthesis in healthy young men.
Collapse
|
9
|
Otto S, Lütjohann D, Kerksiek A, Friedrichs S, Christian Schulze P, Möbius-Winkler S, Pörner TC, Weingärtner O. Increased cholesterol absorption is associated with In-stent-restenosis after stent implantation for stable coronary artery disease. Steroids 2022; 187:109079. [PMID: 35835203 DOI: 10.1016/j.steroids.2022.109079] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Blood cholesterol levels are regulated by competing mechanisms of cholesterol synthesis, absorption and excretion. Plant sterols are natural constituents of plants, are not synthesized in humans, and serve as markers for cholesterol absorption. Ezetimibe lowers the intestinal absorption of cholesterol and plant sterols. We analyzed the associations of differences in cholesterol metabolism, in particular increased cholesterol absorption, and the occurrence of in-stent restenosis (ISR) in patients with stable coronary artery disease. METHODS Elective stent implantation of de novo stenosis was conducted in 59 patients (74.6 % males, 67.2 ± 9.6 years). Cholesterol and non-cholesterol sterols were quantified in serum samples by gas chromatography or mass spectrometry. ISR was assessed by optical coherence tomography (OCT) and quantitative angiography (QCA) after six months. RESULTS Markers for cholesterol absorption (e.g. campesterol-to-cholesterol) were positively associated with ISR measured by QCA (%diameter stenosis, late lumen loss) and OCT (proliferation volume, %area stenosis), whereas markers for cholesterol synthesis (e.g. lathosterol-to-cholesterol) were negatively associated with ISR (%area stenosis: r = -0.271, p = 0.043). There was no association between ISR and total cholesterol, LDL, HDL, triglycerides. Markers for cholesterol absorption (e.g. campesterol-to-cholesterol) were significantly lower in ezetimibe-treated patients compared to patients on a statin only (1.29 ± 0.69 vs. 2.22 ± 1.23; p = 0.007). Combined lipid-lowering with ezetimibe plus statin reduced ISR compared to statin only (13.7 ± 10.4 vs. 22.5 ± 12.1 %diameter stenosis, p = 0.015). CONCLUSIONS Differences in cholesterol metabolism, more specifically increased cholesterol absorption, are associated with ISR.
Collapse
Affiliation(s)
- Sylvia Otto
- Department of Internal Medicine I, Division of Cardiology, Pneumology, Angiology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Silvia Friedrichs
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Paul Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Pneumology, Angiology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Germany
| | - Sven Möbius-Winkler
- Department of Internal Medicine I, Division of Cardiology, Pneumology, Angiology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Germany
| | - Tudor C Pörner
- Asklepios Klinik Wandsbek, Department of Internal Medicine - Cardiology & Pneumology, Hamburg, Germany
| | - Oliver Weingärtner
- Department of Internal Medicine I, Division of Cardiology, Pneumology, Angiology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Germany.
| |
Collapse
|
10
|
Krawczyk M, Niewiadomska O, Jankowska I, Jankowski K, Więckowski S, Lebensztejn D, Więcek S, Gozdowska J, Kułaga Z, Weber SN, Lütjohann D, Lammert F, Socha P. Common variant p.D19H of the hepatobiliary sterol transporter ABCG8 increases the risk of gallstones in children. Liver Int 2022; 42:1585-1592. [PMID: 35129276 DOI: 10.1111/liv.15186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/12/2021] [Accepted: 01/01/2022] [Indexed: 02/13/2023]
Abstract
INTRODUCTION Gallstones are increasingly common in children. Genetic analyses of adult cohorts demonstrated that the sterol transporter ABCG8 p.D19H and Gilbert UGT1A1*28 variants enhance the odds of developing gallstones. The genetic background of common lithiasis in children remains unknown. METHODS Overall, 214 children with gallstone disease (1 month-17 years, 107 boys) were inclueded. The control cohorts comprised 214 children (age 6-17 years, 115 boys) and 172 adults (age 40-92 years, 70 men) without gallstones. The ABCG8 p.D19H and UGT1A1*28 polymorphisms as well as ABCB4 (c.504C>T rs1202283, c.711A>T rs2109505) and NPC1L1 variants (p.V1296V rs217434, c.-18C>A rs41279633) were genotyped using TaqMan assays. Serum concentrations of plant sterols and cholesterol precursors were measured by gas chromatography/mass spectrometry. RESULTS The ABCG8 risk allele was associated with an increased risk of stones (OR = 1.82, p = .03). Children carrying the p.19H allele presented with lower serum concentrations of surrogate markers of intestinal cholesterol absorption and decreased ratios of phytosterols to the cholesterol precursor desmosterol. Carriers of the common NPC1L1 rs217434 allele had an increased gallstone risk compared with stone-free adults (OR 1.90, p < .01). This variant also affected the ratio of phytosterols to cholesterol precursors (p = .03). Other tested variants were not associated with gallstone risk. CONCLUSIONS The p.D19H ABCG8 and, to a lesser extent, NPC1L1 rs217434 variants increase the risk of early-onset gallstone formation. These results point to the presence of a common lithogenic pathway in children and adults.
Collapse
Affiliation(s)
- Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Olga Niewiadomska
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Irena Jankowska
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Krzysztof Jankowski
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Sebastian Więckowski
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Dariusz Lebensztejn
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Bialystok, Bialystok, Poland
| | - Sabina Więcek
- Department of Pediatrics, Silesian Medical Academy, Katowice, Poland
| | - Jolanta Gozdowska
- Department of Transplantation Medicine and Nephrology, Medical University of Warsaw, Warsaw, Poland
| | - Zbigniew Kułaga
- Public Health Department of the Children's Memorial Health Institute, Warsaw, Poland
| | - Susanne N Weber
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Hannover Health Science Campus, Hannover Medical School (MHH), Hannover, Germany
| | - Piotr Socha
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| |
Collapse
|
11
|
Malacarne PF, Ratiu C, Gajos-Draus A, Müller N, Lopez M, Pflüger-Müller B, Ding X, Warwick T, Oo J, Siragusa M, Angioni C, Günther S, Weigert A, Geißlinger G, Lütjohann D, Schunck WH, Fleming I, Brandes RP, Rezende F. Loss of Endothelial Cytochrome P450 Reductase Induces Vascular Dysfunction in Mice. Hypertension 2022; 79:1216-1226. [PMID: 35354305 DOI: 10.1161/hypertensionaha.121.18752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND POR (cytochrome P450 reductase) provides electrons for the catalytic activity of the CYP (cytochrome P450) monooxygenases. CYPs are dual-function enzymes as they generate protective vasoactive mediators derived from polyunsaturated fatty acids but also reactive oxygen species. It is not known in which conditions the endothelial POR/CYP system is beneficial versus deleterious. Here, the activity of all CYP enzymes was eliminated in the vascular endothelium to examine its impact on vascular function. METHODS An endothelial-specific, tamoxifen-inducible POR knockout mouse (ecPOR-/-) was generated. Vascular function was studied by organ chamber experiments. eNOS (endothelial nitric oxide synthase) activity was accessed by heavy arginine/citrulline LC-MS/MS detection and phosphorylation of serine1177 in aortic rings. CYP-derived epoxyeicosatrienoic acids and prostanoids were measured by LC-MS/MS. Gene expression of aorta and endothelial cells was profiled by RNA sequencing. Blood pressure was measured by telemetry. RESULTS Acetylcholine-induced endothelium-dependent relaxation was attenuated in isolated vessels of ecPOR-/- as compared with control mice. Additionally, ecPOR-/- mice had attenuated eNOS activity and eNOS/AKT phosphorylation. POR deletion reduced endothelial stores of CYP-derived epoxyeicosatrienoic acids but increased vascular prostanoids. This phenomenon was paralleled by the induction of genes implicated in eicosanoid generation. In response to Ang II (angiotensin II) infusion, blood pressure increased significantly more in ecPOR-/- mice. Importantly, the cyclooxygenase inhibitor Naproxen selectively lowered the Ang II-induced hypertension in ecPOR-/- mice. CONCLUSIONS POR expression in endothelial cells maintains eNOS activity and its loss results in an overactivation of the vasoconstrictor prostanoid system. Through these mechanisms, loss of endothelial POR induces vascular dysfunction and hypertension.
Collapse
Affiliation(s)
- Pedro Felipe Malacarne
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Corina Ratiu
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Anna Gajos-Draus
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,National Science Centre, Poland (A.G.-D.)
| | - Niklas Müller
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Melina Lopez
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Beatrice Pflüger-Müller
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson (X.D.)
| | - Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - James Oo
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Mauro Siragusa
- Institute for Vascular Signalling, Goethe-University, Frankfurt, Germany. (M.S., I.F.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Carlo Angioni
- Institute for Clinical Pharmacology, Goethe-University, Frankfurt, Germany. (C.A., G.G.)
| | - Stefan Günther
- Institute for Heart and Lung Research, Max Planck Institute, Bad Nauheim, Germany (S.G.)
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe-University, Frankfurt, Germany. (A.W.)
| | - Gerd Geißlinger
- Institute for Clinical Pharmacology, Goethe-University, Frankfurt, Germany. (C.A., G.G.)
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Pharmacology, University of Bonn, Germany (D.L.)
| | | | - Ingrid Fleming
- Institute for Vascular Signalling, Goethe-University, Frankfurt, Germany. (M.S., I.F.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| | - Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany. (P.F.M., C.R., A.G.-D., N.M., M.L., B.P.-M., T.W., J.O., R.P.B., F.R.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt, Germany (P.F.M., C.R., N.M., M.L., B.P.-M., T.W., J.O., M.S., I.F., R.P.B., F.R.)
| |
Collapse
|
12
|
Ghaleb Y, Elbitar S, Philippi A, El Khoury P, Azar Y, Andrianirina M, Loste A, Abou-Khalil Y, Nicolas G, Le Borgne M, Moulin P, Di-Filippo M, Charrière S, Farnier M, Yelnick C, Carreau V, Ferrières J, Lecerf JM, Derksen A, Bernard G, Gauthier MS, Coulombe B, Lütjohann D, Fin B, Boland A, Olaso R, Deleuze JF, Rabès JP, Boileau C, Abifadel M, Varret M. Whole Exome/Genome Sequencing Joint Analysis of a Family with Oligogenic Familial Hypercholesterolemia. Metabolites 2022; 12:metabo12030262. [PMID: 35323704 PMCID: PMC8955453 DOI: 10.3390/metabo12030262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
Autosomal Dominant Hypercholesterolemia (ADH) is a genetic disorder caused by pathogenic variants in LDLR, APOB, PCSK9 and APOE genes. We sought to identify new candidate genes responsible for the ADH phenotype in patients without pathogenic variants in the known ADH-causing genes by focusing on a French family with affected and non-affected members who presented a high ADH polygenic risk score (wPRS). Linkage analysis, whole exome and whole genome sequencing resulted in the identification of variants p.(Pro398Ala) in CYP7A1, p.(Val1382Phe) in LRP6 and p.(Ser202His) in LDLRAP1. A total of 6 other variants were identified in 6 of 160 unrelated ADH probands: p.(Ala13Val) and p.(Aps347Asn) in CYP7A1; p.(Tyr972Cys), p.(Thr1479Ile) and p.(Ser1612Phe) in LRP6; and p.(Ser202LeufsTer19) in LDLRAP1. All six probands presented a moderate wPRS. Serum analyses of carriers of the p.(Pro398Ala) variant in CYP7A1 showed no differences in the synthesis of bile acids compared to the serums of non-carriers. Functional studies of the four LRP6 mutants in HEK293T cells resulted in contradictory results excluding a major effect of each variant alone. Within the family, none of the heterozygous for only the LDLRAP1 p.(Ser202His) variant presented ADH. Altogether, each variant individually does not result in elevated LDL-C; however, the oligogenic combination of two or three variants reveals the ADH phenotype.
Collapse
Affiliation(s)
- Youmna Ghaleb
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé (PTS), Saint-Joseph University, Beirut 1004 2020, Lebanon
| | - Sandy Elbitar
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé (PTS), Saint-Joseph University, Beirut 1004 2020, Lebanon
| | - Anne Philippi
- Institut Cochin, Bâtiment Faculté Inserm U1016, Cnrs UMR8104, Université de Paris Faculté de Médecine, F-75014 Paris, France;
| | - Petra El Khoury
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé (PTS), Saint-Joseph University, Beirut 1004 2020, Lebanon
| | - Yara Azar
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé (PTS), Saint-Joseph University, Beirut 1004 2020, Lebanon
- Laboratory for Vascular Translational Science, Paris Cité University, Sorbonne Paris Nord University, F-75013 Paris, France;
| | - Miangaly Andrianirina
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
| | - Alexia Loste
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory for Vascular Translational Science, Paris Cité University, Sorbonne Paris Nord University, F-75013 Paris, France;
| | - Yara Abou-Khalil
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé (PTS), Saint-Joseph University, Beirut 1004 2020, Lebanon
- Laboratory for Vascular Translational Science, Paris Cité University, Sorbonne Paris Nord University, F-75013 Paris, France;
| | - Gaël Nicolas
- Laboratory for Vascular Translational Science, Paris Cité University, Sorbonne Paris Nord University, F-75013 Paris, France;
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l’Inflammation, F-75018 Paris, France
| | - Marie Le Borgne
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory for Vascular Translational Science, Paris Cité University, Sorbonne Paris Nord University, F-75013 Paris, France;
| | - Philippe Moulin
- Department of Endocrinology, Nutrition and Metabolic Diseases, Hospices Civils de Lyon, Louis Pradel Cardiovascular Hospital, F-69500 Bron, France; (P.M.); (S.C.)
- CarMen Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, F-69921 Oullins, France;
| | - Mathilde Di-Filippo
- CarMen Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, F-69921 Oullins, France;
- Hospices Civils de Lyon, Department of Biochemistry and Molecular Biology, F-69500 Bron, France
| | - Sybil Charrière
- Department of Endocrinology, Nutrition and Metabolic Diseases, Hospices Civils de Lyon, Louis Pradel Cardiovascular Hospital, F-69500 Bron, France; (P.M.); (S.C.)
- CarMen Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, F-69921 Oullins, France;
| | - Michel Farnier
- EA 7460 Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne-Franche Comté, F-21078 Dijon, France;
| | - Cécile Yelnick
- Département de Médecine Interne et Immunologie Clinique Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO) CHU de Lille, F-59037 Lille, France;
- U1167 Risk Factors and Molecular Determinants of Aging-Related Diseases, Inserm CHU de Lille, Lille University, F-59000 Lille, France
| | - Valérie Carreau
- Department of Endocrinology and Prevention of Cardiovascular Disease, Institute of Cardio Metabolism and Nutrition (ICAN), La Pitié-Salpêtrière Hospital, AP-HP, F-75005 Paris, France;
| | - Jean Ferrières
- Department of Cardiology, Toulouse Rangueil University Hospital, UMR 1295 INSERM, F-31400 Toulouse, France;
| | - Jean-Michel Lecerf
- Nutrition Department, Institut Pasteur de Lille, CEDEX, F-59019 Lille, France;
| | - Alexa Derksen
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC H3A 0G4, Canada; (A.D.); (G.B.)
- Translational Proteomics Laboratory, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; (M.-S.G.); (B.C.)
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC H3A 0G4, Canada
| | - Geneviève Bernard
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC H3A 0G4, Canada; (A.D.); (G.B.)
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC H3A 0G4, Canada
- Department of Pediatrics, McGill University, Montréal, QC H3A 0G4, Canada
- Department of Human Genetics, McGill University, Montréal, QC H3A 0G4, Canada
- Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Marie-Soleil Gauthier
- Translational Proteomics Laboratory, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; (M.-S.G.); (B.C.)
| | - Benoit Coulombe
- Translational Proteomics Laboratory, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; (M.-S.G.); (B.C.)
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, F-53127 Bonn, Germany;
| | - Bertrand Fin
- CEA, Centre National de Recherche en Génomique Humaine, Laboratory of Excellence GENMED (Medical Genomics), Paris-Saclay University, F-91057 Evry, France; (B.F.); (A.B.); (R.O.); (J.-F.D.)
| | - Anne Boland
- CEA, Centre National de Recherche en Génomique Humaine, Laboratory of Excellence GENMED (Medical Genomics), Paris-Saclay University, F-91057 Evry, France; (B.F.); (A.B.); (R.O.); (J.-F.D.)
| | - Robert Olaso
- CEA, Centre National de Recherche en Génomique Humaine, Laboratory of Excellence GENMED (Medical Genomics), Paris-Saclay University, F-91057 Evry, France; (B.F.); (A.B.); (R.O.); (J.-F.D.)
| | - Jean-François Deleuze
- CEA, Centre National de Recherche en Génomique Humaine, Laboratory of Excellence GENMED (Medical Genomics), Paris-Saclay University, F-91057 Evry, France; (B.F.); (A.B.); (R.O.); (J.-F.D.)
- Centre d’Etude du Polymorphisme Humain, Fondation Jean Dausset, F-75019 Paris, France
| | - Jean-Pierre Rabès
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Department of Biochemistry and Molecular Genetics, Ambroise Paré University Hospital (APHP), Université Paris-Saclay, F-92104 Boulogne-Billancourt, France
- UFR (Unite de Formation et de Recherche) Simone Veil-Santé, Versailles-Saint-Quentin-en-Yvelines University, F-78180 Montigny-le-Bretonneux, France
| | - Catherine Boileau
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory for Vascular Translational Science, Paris Cité University, Sorbonne Paris Nord University, F-75013 Paris, France;
- Genetic Department, AP-HP, Hôpital Bichat, F-75018 Paris, France
| | - Marianne Abifadel
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé (PTS), Saint-Joseph University, Beirut 1004 2020, Lebanon
| | - Mathilde Varret
- INSERM, Laboratory for Vascular Translational Science (LVTS), F-75018 Paris, France; (Y.G.); (S.E.); (P.E.K.); (Y.A.); (M.A.); (A.L.); (Y.A.-K.); (M.L.B.); (J.-P.R.); (C.B.); (M.A.)
- Laboratory for Vascular Translational Science, Paris Cité University, Sorbonne Paris Nord University, F-75013 Paris, France;
- Correspondence: ; Tel.: +33-1402-57521
| |
Collapse
|
13
|
Baumgartner S, Lütjohann D, Husche C, Kerksiek A, Groen AK, Mensink RP, Plat J. Plasma oxyphytosterols most likely originate from hepatic oxidation and subsequent spill-over in the circulation. J Steroid Biochem Mol Biol 2022; 216:106039. [PMID: 34861389 DOI: 10.1016/j.jsbmb.2021.106039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
We evaluated oxyphytosterol (OPS) concentrations in plasma and various tissues of two genetically modified mouse models with either increased cholesterol (apoE KO mice) or increased cholesterol and plant sterol (PS) concentrations (apoExABCG8 dKO mice). Sixteen female apoE KO and 16 dKO mice followed the same standard, low OPS-chow diet. Animals were euthanized at 36 weeks to measure PS and OPS concentrations in plasma, brain, liver and aortic tissue. Cholesterol and oxysterol (OS) concentrations were analyzed as reference for sterol oxidation in general. Plasma campesterol (24.1 ± 4.3 vs. 11.8 ± 3.0 mg/dL) and sitosterol (67.4 ± 12.7 vs. 4.9 ± 1.1 mg/dL) concentrations were severely elevated in the dKO compared to the apoE KO mice (p < 0.001). Also, in aortic and brain tissue, PS levels were significantly elevated in dKO. However, plasma, aortic and brain OPS concentrations were comparable or even lower in the dKO mice. In contrast, in liver tissue, both PS and OPS concentrations were severely elevated in the dKO compared to apoE KO mice (sum OPS: 7.4 ± 1.6 vs. 4.1 ± 0.8 ng/mg, p < 0.001). OS concentrations followed cholesterol concentrations in plasma and all tissues suggesting ubiquitous oxidation. Despite severely elevated PS concentrations, OPS concentrations were only elevated in liver tissue, suggesting that OPS are primarily formed in the liver and plasma concentrations originate from hepatic spill-over into the circulation.
Collapse
Affiliation(s)
- S Baumgartner
- Department of Nutrition and Movement Sciences. NUTRIM School of Nutrition and Translational Research in Metabolism. Maastricht University, Maastricht, 6200 MD, The Netherlands.
| | - D Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, D-53127, Germany
| | - C Husche
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, D-53127, Germany
| | - A Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, D-53127, Germany
| | - A K Groen
- Amsterdam Diabetes Center and Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, 1105 AZ, The Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 ZG, The Netherlands
| | - R P Mensink
- Department of Nutrition and Movement Sciences. NUTRIM School of Nutrition and Translational Research in Metabolism. Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - J Plat
- Department of Nutrition and Movement Sciences. NUTRIM School of Nutrition and Translational Research in Metabolism. Maastricht University, Maastricht, 6200 MD, The Netherlands
| |
Collapse
|
14
|
Spohner AK, Jakobi K, Trautmann S, Thomas D, Schumacher F, Kleuser B, Lütjohann D, El-Hindi K, Grösch S, Pfeilschifter J, Saba JD, Meyer zu Heringdorf D. Mouse Liver Compensates Loss of Sgpl1 by Secretion of Sphingolipids into Blood and Bile. Int J Mol Sci 2021; 22:10617. [PMID: 34638955 PMCID: PMC8508615 DOI: 10.3390/ijms221910617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
Sphingosine 1 phosphate (S1P) lyase (Sgpl1) catalyses the irreversible cleavage of S1P and thereby the last step of sphingolipid degradation. Loss of Sgpl1 in humans and mice leads to accumulation of sphingolipids and multiple organ injuries. Here, we addressed the role of hepatocyte Sgpl1 for regulation of sphingolipid homoeostasis by generating mice with hepatocyte-specific deletion of Sgpl1 (Sgpl1HepKO mice). Sgpl1HepKO mice had normal body weight, liver weight, liver structure and liver enzymes both at the age of 8 weeks and 8 months. S1P, sphingosine and ceramides, but not glucosylceramides or sphingomyelin, were elevated by ~1.5-2-fold in liver, and this phenotype did not progress with age. Several ceramides were elevated in plasma, while plasma S1P was normal. Interestingly, S1P and glucosylceramides, but not ceramides, were elevated in bile of Sgpl1HepKO mice. Furthermore, liver cholesterol was elevated, while LDL cholesterol decreased in 8-month-old mice. In agreement, the LDL receptor was upregulated, suggesting enhanced uptake of LDL cholesterol. Expression of peroxisome proliferator-activated receptor-γ, liver X receptor and fatty acid synthase was unaltered. These data show that mouse hepatocytes largely compensate the loss of Sgpl1 by secretion of accumulating sphingolipids in a specific manner into blood and bile, so that they can be excreted or degraded elsewhere.
Collapse
Affiliation(s)
- Anna Katharina Spohner
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| | - Katja Jakobi
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| | - Sandra Trautmann
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Dominique Thomas
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Fabian Schumacher
- Institut für Pharmazie, Pharmakologie und Toxikologie, Freie Universität Berlin, Königin-Luise-Straße 2-4, 14195 Berlin, Germany; (F.S.); (B.K.)
| | - Burkhard Kleuser
- Institut für Pharmazie, Pharmakologie und Toxikologie, Freie Universität Berlin, Königin-Luise-Straße 2-4, 14195 Berlin, Germany; (F.S.); (B.K.)
| | - Dieter Lütjohann
- Institut für Klinische Chemie und Pharmakologie, Universitätsklinikum Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany;
| | - Khadija El-Hindi
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Sabine Grösch
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| | - Julie D. Saba
- Department of Pediatrics, Division of Hematology/Oncology, University of California, 505 Parnassus Ave, San Francisco, CA 94143, USA;
| | - Dagmar Meyer zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| |
Collapse
|
15
|
Houben T, Yadati T, de Kruijf R, Gijbels MJJ, Luiken JJFP, van Zandvoort M, Kapsokalyvas D, Lütjohann D, Westerterp M, Plat J, Leake D, Shiri-Sverdlov R. Pro-Inflammatory Implications of 2-Hydroxypropyl-β-cyclodextrin Treatment. Front Immunol 2021; 12:716357. [PMID: 34489968 PMCID: PMC8417873 DOI: 10.3389/fimmu.2021.716357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/23/2021] [Indexed: 11/24/2022] Open
Abstract
Lifestyle- and genetically induced disorders related to disturbances in cholesterol metabolism have shown the detrimental impact of excessive cholesterol levels on a plethora of pathological processes such as inflammation. In this context, two-hydroxypropyl-β-cyclodextrin (CD) is increasingly considered as a novel pharmacological compound to decrease cellular cholesterol levels due to its ability to increase cholesterol solubility. However, recent findings have reported contra-indicating events after the use of CD questioning the clinical applicability of this compound. Given its potential as a therapeutic compound in metabolic inflammatory diseases, in this study, we evaluated the inflammatory effects of CD administration in the context of cholesterol-induced metabolic inflammation in vivo and in vitro. The inflammatory and cholesterol-depleting effects of CD were first investigated in low-density lipoprotein receptor knockout (Ldlr-/) mice that were transplanted with Npc1nih or Npc1wt bone marrow and were fed either regular chow or a high-fat, high-cholesterol (HFC) diet for 12 weeks, thereby creating an extreme model of lysosomal cholesterol-induced metabolic inflammation. In the final three weeks, these mice received daily injections of either control (saline) or CD subcutaneously. Subsequently, the inflammatory properties of CD were investigated in vitro in two macrophage cell lines and in murine bone marrow-derived macrophages (BMDMs). While CD administration improved cholesterol mobilization outside lysosomes in BMDMs, an overall pro-inflammatory profile was observed after CD treatment, evidenced by increased hepatic inflammation in vivo and a strong increase in cytokine release and inflammatory gene expression in vitro in murine BMDMs and macrophages cell lines. Nevertheless, this CD-induced pro-inflammatory profile was time-dependent, as short term exposure to CD did not result in a pro-inflammatory response in BMDM. While CD exerts desired cholesterol-depleting effects, its inflammatory effect is dependent on the exposure time. As such, using CD in the clinic, especially in a metabolic inflammatory context, should be closely monitored as it may lead to undesired, pro-inflammatory side effects.
Collapse
Affiliation(s)
- Tom Houben
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands
| | - Tulasi Yadati
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands
| | - Robbin de Kruijf
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands
| | - Marion J J Gijbels
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands
| | - Joost J F P Luiken
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands
| | - Marc van Zandvoort
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands.,School for Oncology and Developmental Biology GROW, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases CARIM Maastricht University, Maastricht, Netherlands.,Institute for Molecular Cardiovascular Research IMCAR, Rheinisch-Westfälische Technische Hogeschool (RWTH) Aachen University, Aachen, Germany
| | - Dimitris Kapsokalyvas
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands.,School for Oncology and Developmental Biology GROW, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases CARIM Maastricht University, Maastricht, Netherlands.,Institute for Molecular Cardiovascular Research IMCAR, Rheinisch-Westfälische Technische Hogeschool (RWTH) Aachen University, Aachen, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Marit Westerterp
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| | - David Leake
- School of Biological Sciences, University of Reading, Health and Life Sciences Building, Whiteknights, Reading, United Kingdom
| | - Ronit Shiri-Sverdlov
- Departments of Genetics and Cell Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht, Maastricht, Netherlands
| |
Collapse
|
16
|
Emrich IE, Heine GH, Schulze PC, Rogacev KS, Fliser D, Wagenpfeil S, Böhm M, Lütjohann D, Weingärtner O. Markers of cholesterol synthesis to cholesterol absorption across the spectrum of non-dialysis CKD: An observational study. Pharmacol Res Perspect 2021; 9:e00801. [PMID: 34128357 PMCID: PMC8204094 DOI: 10.1002/prp2.801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
In dialysis patients, cholesterol-lowering therapy with statins is less effective than in other high-risk patients. This may be explained by a shift from cholesterol synthesis toward cholesterol absorption. In line, markers of cholesterol absorption-such as campesterol-better predict atherosclerotic cardiovascular events than markers of cholesterol synthesis-such as lathosterol-in dialysis patients. To test the association between markers of cholesterol absorption such as campesterol-and markers of cholesterol synthesis-such as lathosterol-against cardiovascular events in non-dialysis CKD patients. Altogether 251 patients those not on lipid-lowering agents were followed annually for the composite endpoint atherosclerotic cardiovascular disease (ASCVD) and all-cause death. During follow-up of 5.2 ± 2.1 years, 61 participants reached the primary endpoint atherosclerotic cardiovascular disease/all-cause death [ASCVD/D], 47 participants suffered from ASCVD, and 46 participants died. In univariate Cox regression analysis, campesterol/lathosterol ratio did not significantly predict ASCVD/D (HR 0.643; 0.358-1.155; 3rd vs. 1st tertile), all-cause death (HR 1.309; 0.604-2.838; 3rd vs. 1st tertile) nor ASCVD (HR 0.589; 0.311-1.118; 3rd vs. 1st tertile). We did not observe a shift from cholesterol synthesis to cholesterol absorption across the spectrum of non-dialysis CKD. Campesterol/lathosterol ratio did not predict future ASCVD or all-cause death in non-dialysis CKD.
Collapse
Affiliation(s)
- Insa E. Emrich
- Internal Medicine III – Cardiology, Angiology and Intensive Care MedicineSaarland University Medical CenterSaarland UniversityHomburgGermany
| | | | - P. Christian Schulze
- Department of Internal Medicine I – Cardiology, Angiology, Intensive Care MedicineUniversity Hospital JenaJenaGermany
| | - Kyrill S. Rogacev
- Internal Medicine II – CardiologySana Hanse‐Klinikum WismarWismarGermany
| | - Danilo Fliser
- Internal Medicine IV – Nephrology and HypertensionSaarland University Medical CenterHomburgGermany
| | - Stefan Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical InformaticsSaarland UniversityUniversity Medical CenterHomburgGermany
| | - Michael Böhm
- Internal Medicine III – Cardiology, Angiology and Intensive Care MedicineSaarland University Medical CenterSaarland UniversityHomburgGermany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical PharmacologyMedical FacultyUniversity of BonnBonnGermany
| | - Oliver Weingärtner
- Department of Internal Medicine I – Cardiology, Angiology, Intensive Care MedicineUniversity Hospital JenaJenaGermany
- School of Medicine and Health SciencesCarl von Ossietzky UniversityUniversity Clinic of Internal Medicine, CardiologyOldenburgGermany
| |
Collapse
|
17
|
Christidis G, Karatayli E, Hall RA, Weber SN, Reichert MC, Hohl M, Qiao S, Boehm U, Lütjohann D, Lammert F, Karatayli SC. Fibroblast Growth Factor 21 Response in a Preclinical Alcohol Model of Acute-on-Chronic Liver Injury. Int J Mol Sci 2021; 22:7898. [PMID: 34360670 PMCID: PMC8348955 DOI: 10.3390/ijms22157898] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS Fibroblast growth factor (FGF) 21 has recently been shown to play a potential role in bile acid metabolism. We aimed to investigate the FGF21 response in an ethanol-induced acute-on-chronic liver injury (ACLI) model in Abcb4-/- mice with deficiency of the hepatobiliary phospholipid transporter. METHODS Total RNA was extracted from wild-type (WT, C57BL/6J) and Abcb4-/- (KO) mice, which were either fed a control diet (WT-Cont and KO-Cont groups; n = 28/group) or ethanol diet, followed by an acute ethanol binge (WT-EtOH and KO-EtOH groups; n = 28/group). A total of 58 human subjects were recruited into the study, including patients with alcohol-associated liver disease (AALD; n = 31) and healthy controls (n = 27). The hepatic and ileal expressions of genes involved in bile acid metabolism, plasma FGF levels, and bile acid and its precursors 7α- and 27-hydroxycholesterol (7α- and 27-OHC) concentrations were determined. Primary mouse hepatocytes were isolated for cell culture experiments. RESULTS Alcohol feeding significantly induced plasma FGF21 and decreased hepatic Cyp7a1 levels. Hepatic expression levels of Fibroblast growth factor receptor 1 (Fgfr1), Fgfr4, Farnesoid X-activated receptor (Fxr), and Small heterodimer partner (Shp) and plasma FGF15/FGF19 levels did not differ with alcohol challenge. Exogenous FGF21 treatment suppressed Cyp7a1 in a dose-dependent manner in vitro. AALD patients showed markedly higher FGF21 and lower 7α-OHC plasma levels while FGF19 did not differ. CONCLUSIONS The simultaneous upregulation of FGF21 and downregulation of Cyp7a1 expressions upon chronic plus binge alcohol feeding together with the invariant plasma FGF15 and hepatic Shp and Fxr levels suggest the presence of a direct regulatory mechanism of FGF21 on bile acid homeostasis through inhibition of CYP7A1 by an FGF15-independent pathway in this ACLI model. Lay Summary: Alcohol challenge results in the upregulation of FGF21 and repression of Cyp7a1 expressions while circulating FGF15 and hepatic Shp and Fxr levels remain constant both in healthy and pre-injured livers, suggesting the presence of an alternative FGF15-independent regulatory mechanism of FGF21 on bile acid homeostasis through the inhibition of Cyp7a1.
Collapse
Affiliation(s)
- Grigorios Christidis
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (G.C.); (E.K.); (R.A.H.); (S.N.W.); (M.C.R.); (F.L.)
| | - Ersin Karatayli
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (G.C.); (E.K.); (R.A.H.); (S.N.W.); (M.C.R.); (F.L.)
| | - Rabea A. Hall
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (G.C.); (E.K.); (R.A.H.); (S.N.W.); (M.C.R.); (F.L.)
| | - Susanne N. Weber
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (G.C.); (E.K.); (R.A.H.); (S.N.W.); (M.C.R.); (F.L.)
| | - Matthias C. Reichert
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (G.C.); (E.K.); (R.A.H.); (S.N.W.); (M.C.R.); (F.L.)
| | - Mathias Hohl
- Department of Medicine III, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany;
| | - Sen Qiao
- Department of Pharmacology and Toxicology, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (S.Q.); (U.B.)
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (S.Q.); (U.B.)
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (G.C.); (E.K.); (R.A.H.); (S.N.W.); (M.C.R.); (F.L.)
- Hannover Health Sciences Campus, Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Senem Ceren Karatayli
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (G.C.); (E.K.); (R.A.H.); (S.N.W.); (M.C.R.); (F.L.)
| |
Collapse
|
18
|
Frank AC, Raue R, Fuhrmann DC, Sirait-Fischer E, Reuse C, Weigert A, Lütjohann D, Hiller K, Syed SN, Brüne B. Lactate dehydrogenase B regulates macrophage metabolism in the tumor microenvironment. Am J Cancer Res 2021; 11:7570-7588. [PMID: 34158867 PMCID: PMC8210612 DOI: 10.7150/thno.58380] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/04/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Glucose metabolism in the tumor-microenvironment is a fundamental hallmark for tumor growth and intervention therein remains an attractive option for anti-tumor therapy. Whether tumor-derived factors such as microRNAs (miRs) regulate glucose metabolism in stromal cells, especially in tumor-associated macrophages (TAMs), to hijack them for trophic support, remains elusive. Methods: Ago-RIP-Seq identified macrophage lactate dehydrogenase B (LDHB) as a target of tumor-derived miR-375 in both 2D/3D cocultures and in murine TAMs from a xenograft mouse model. The prognostic value was analyzed by ISH and multiplex IHC of breast cancer patient tissues. Functional consequences of the miR-375-LDHB axis in TAMs were investigated upon mimic/antagomir treatment by live metabolic flux assays, GC/MS, qPCR, Western blot, lentiviral knockdown and FACS. The therapeutic potential of a combinatorial miR-375-decoy/simvastatin treatment was validated by live cell imaging. Results: Macrophage LDHB decreased in murine and human breast carcinoma. LDHB downregulation increase aerobic glycolysis and lactagenesis in TAMs in response to tumor-derived miR-375. Lactagenesis reduced fatty acid synthesis but activated SREBP2, which enhanced cholesterol biosynthesis in macrophages. LDHB downregulation skewed TAMs to function as a lactate and sterol/oxysterol source for the proliferation of tumor cells. Restoring of LDHB expression potentiated inhibitory effects of simvastatin on tumor cell proliferation. Conclusion: Our findings identified a crucial role of LDHB in macrophages and established tumor-derived miR-375 as a novel regulator of macrophage metabolism in breast cancer, which might pave the way for strategies of combinatorial cancer cell/stroma cell interventions.
Collapse
|
19
|
Magro dos Reis I, Houben T, Gijbels MJJ, Lütjohann D, Plat J, Shiri-Sverdlov R. Anti-Inflammatory Effects of Dietary Plant Stanol Supplementation Are Largely Dependent on the Intake of Cholesterol in a Mouse Model of Metabolic Inflammation. Biomedicines 2021; 9:biomedicines9050518. [PMID: 34066407 PMCID: PMC8148209 DOI: 10.3390/biomedicines9050518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 01/25/2023] Open
Abstract
The prevalence of metabolic disorders characterized by chronic inflammation has been on a sharp rise for decades. As such, tools that address metabolic and inflammatory dysregulation are of great importance. Plant stanols are well-known for reducing intestinal cholesterol absorption and may also have direct anti-inflammatory effects. In this study, our aim was to investigate to what extent the benefits of dietary plant stanol supplementation depend on dietary cholesterol intake in an experimental mouse model for cholesterol-induced metabolic inflammation. Here, we used Ldlr−/− mice transplanted with Npc1nih-derived bone marrow, featuring feature bone marrow-derived immune cells characterized by chronic inflammation induced by lysosomal lipid accumulation. Npc1nih- and Npc1wt-transplanted mice were placed on either a high fat, high cholesterol (HFC) or on a chow diet low in cholesterol, with or without 2% plant stanols supplementation. At the end of the study, the metabolic and inflammatory status of the mice was analyzed. Plant stanol supplementation to the HFC diet reduced liver cholesterol levels and improved lipid metabolism and liver inflammation, particularly in Npc1nih-tp mice. In contrast, plant stanol supplementation to the chow diet did not significantly improve the aforementioned parameters, though similar reductive trends to those in the HFC diet setting were observed regarding liver cholesterol accumulation and liver inflammatory markers. The effects of dietary plant stanol supplementation on dietary cholesterol-induced inflammation are largely dependent on dietary cholesterol intake. Future research should verify whether other models of metabolic inflammation exhibit similar stanol-related effects on inflammation.
Collapse
Affiliation(s)
- Inês Magro dos Reis
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (I.M.d.R.); (T.H.)
| | - Tom Houben
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (I.M.d.R.); (T.H.)
| | - Marion J. J. Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM,), School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands;
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, Venusberg-Campus 1, University Hospital Bonn, D-53127 Bonn, Germany;
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (I.M.d.R.); (T.H.)
- Correspondence: ; Tel.: +31-433-881-746
| |
Collapse
|
20
|
Lütjohann D, Stellaard F, Bölükbasi B, Kerksiek A, Parhofer KG, Laufs U. Anti-PCSK 9 antibodies increase the ratios of the brain-specific oxysterol 24S-hydroxycholesterol to cholesterol and to 27-hydroxycholesterol in the serum. Br J Clin Pharmacol 2021; 87:4252-4261. [PMID: 33792095 DOI: 10.1111/bcp.14841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/28/2021] [Accepted: 03/15/2021] [Indexed: 11/27/2022] Open
Abstract
AIMS The serum ratios of the brain-specific oxysterol 24S-hydroxycholesterol (24S-OHC) to cholesterol and to 27-OHC reflect brain cholesterol turnover. We studied the effect of proprotein convertase subtilisin/kexin type 9 monoclonal antibodies (PCSK9ab) that enhance low-density lipoprotein receptor activity on serum cholesterol and oxysterol concentrations. METHODS Twenty-eight hypercholesterolaemic patients (15 males and 13 females) responding insufficiently to maximally tolerated statin and/or ezetimibe therapy were additionally subcutanously treated biweekly with either the PCSK9ab alirocumab (150 mg, n = 13) or evolocumab (140 mg, n = 15). Fasting serum cholesterol was measured by gas chromatography and the oxysterols 24S-OHC and 27-OHC using gas chromatography-mass spectrometry before, after 1-month (n = 28) and after 3-month (n = 13) treatment. RESULTS As expected, PCSK9ab treatment lowered serum cholesterol and oxysterol levels after 1 month. The serum ratio of 24S-OHC to cholesterol increased after 1 month by 17 ± 28% (mean ± standard deviation; 95% confidence interval [CI]: 5.8 to 28%; P < .01) and 24S-OHC to 27-OHC by 15 ± 39% (95% CI: 0.2 to 30%; P < .01). Within 3 months, 24S-OHC to cholesterol increased by 2.8 μg g-1 mo-1 (95% CI: 2.1 to 3.6; P < .01) and 24S-OHC to 27-OHC by 0.019 mo-1 (95% CI: 0.007 to 0.032; P < .01). CONCLUSION The serum ratios of 24S-OHC to cholesterol and to 27-OHC increased after treatment with PCSK9ab. We hypothesize that this is caused by a reduced entrance of 27-OHC into the brain, increased synthesis of brain cholesterol, increased production of 24S-OHC and its secretion across the blood-brain barrier.
Collapse
Affiliation(s)
- Dieter Lütjohann
- Institut für Klinische Chemie und Klinische Pharmakologie, Universitätsklinikum Bonn, Bonn, Germany
| | - Frans Stellaard
- Institut für Klinische Chemie und Klinische Pharmakologie, Universitätsklinikum Bonn, Bonn, Germany
| | - Bediha Bölükbasi
- Klinik für Innere Medizin III (Kardiologie, Angiologie und Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg, Germany
| | - Anja Kerksiek
- Institut für Klinische Chemie und Klinische Pharmakologie, Universitätsklinikum Bonn, Bonn, Germany
| | - Klaus G Parhofer
- Medizinische Klinik IV-Campus Großhadern, Klinikum der Universität München, Munich, Germany
| | - Ulrich Laufs
- Klinik für Innere Medizin III (Kardiologie, Angiologie und Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg, Germany.,Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| |
Collapse
|
21
|
Vodnik VV, Mojić M, Stamenović U, Otoničar M, Ajdžanović V, Maksimović-Ivanić D, Mijatović S, Marković MM, Barudžija T, Filipović B, Milošević V, Šošić-Jurjević B. Development of genistein-loaded gold nanoparticles and their antitumor potential against prostate cancer cell lines. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112078. [PMID: 33947570 DOI: 10.1016/j.msec.2021.112078] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 01/21/2023]
Abstract
Soy isoflavone genistein (Gen) exerts beneficial effects against prostate cancer cells in vitro and in vivo. However, its use as a chemoprevention/therapeutic agent is largely limited due to its low bioavailability. In this study we synthesized two variants of a new delivery system, genistein-gold nanoparticles conjugates Gen@AuNPs1 and Gen@AuNPs2, by an environmentally friendly method, using a dual role of Gen to reduce Au3+ and stabilize the formed AuNPs, with no additional component. The formation of Gen@AuNPs was confirmed via UV-Vis spectroscopy, FTIR, and Raman spectra measurements. The spherical shape and uniform size of Gen@AuNPs1 and Gen@AuNPs2 (10 ± 2 and 23 ± 3 nm, respectively), were determined by transmission electron microscopy. The nano-conjugates also varied in hydrodynamic diameter (65.0 ± 1.7 and 153.0 ± 2.2 nm) but had similar negative zeta potential (-35.0 ± 2.5 and -37.0 ± 1.6 mV), as measured by dynamic light scattering. The Gen loading was estimated to be 46 and 48%, for Gen@AuNPs1 and Gen@AuNPs2, respectively. The antiproliferative activities of GenAuNPs were confirmed by MTT test in vitro on three malignant prostate carcinoma cell lines (PC3, DU 145, and LNCaP), while selectivity toward malignant phenotype was confirmed using non-cancerous MRC-5 cells. Flow cytometric analysis showed that the inhibition on cell proliferation of more potent Gen@AuNPs1 nano-conjugate is comparable with the effects of free Gen. In conclusion, the obtained results, including physicochemical characterization of newly synthesized AuNPs loaded with Gen, cytotoxicity, and IC50 assessments, indicate their stability and bioactivity as an antioxidant and anti-prostate cancer agent, with low toxicity against human primary cells.
Collapse
Affiliation(s)
- Vesna V Vodnik
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Belgrade, Serbia.
| | - Marija Mojić
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Una Stamenović
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Belgrade, Serbia
| | - Mojca Otoničar
- Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Vladimir Ajdžanović
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Sanja Mijatović
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Mirjana M Marković
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Belgrade, Serbia
| | - Tanja Barudžija
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Belgrade, Serbia
| | - Branko Filipović
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Verica Milošević
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Branka Šošić-Jurjević
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| |
Collapse
|
22
|
Ajdžanović V, Miler M, Šošić-Jurjević B, Filipović B, Milenkovic D, Jakovljević V, Milošević V. Soy isoflavone-caused shunting of the corticosteroidogenesis pathways in andropausal subjects: Top-down impulse for the optimal supplementation design. Med Hypotheses 2021; 148:110516. [PMID: 33548764 DOI: 10.1016/j.mehy.2021.110516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/21/2021] [Accepted: 01/24/2021] [Indexed: 11/20/2022]
Abstract
In a series of our previous works, we revealed the beneficial effects of applied soy isoflavones (genistein or daidzein) on the wide context of corticosteroidogenesis in vivo, in a rat model of the andropause. Soy isoflavones decreased the circulating levels of pituitary adrenocorticotropic hormone, inhibited aldosterone secretion, as well as corticosterone production and secretion, but stimulated dehydroepiandrosterone secretion, all in andropausal rats. In vitro studies indicate that the mechanism underlying these hormonal changes relies on inhibition of the pituitary tyrosine kinase and adrenocortical 3β-hydroxysteroid dehydrogenase enzymes by soy isoflavones. Although the clinical studies are in their infancy, the opinion is that genistein and daidzein have therapeutic potential for the safe treatment of ageing-caused androgen deprivation and glucocorticoid excess with related metabolic/hemodynamic issues in males. Our accumulated experience and knowledge in the field of biomedical effects of plant polyphenols have provided a platform for potential recommending the agenda to organize and accelerate experimental research aimed at producing the optimal supplementation. We hypothesize that an in vivo approach should first be exploited in the sequence of investigative steps, followed by in vitro studies and synchronously conducted molecular docking analyses. In vivo research, besides establishing the margin of exposure safety or adjustment of the correct polyphenol dose, enables identification and quantification of the metabolites of applied polyphenols in the blood. Subsequent in vitro exploitation of the metabolites and related docking analyses provide clarification of the molecular mechanisms of action of applied polyphenols. Chemical modification of the polyphenol structure or coupling it with nanoparticles might be the next step in optimizing the design of supplementation. Selected, intact or chemically-modified polyphenol molecules should be included in preclinical studies on a more closely-related species, while clinical studies would finally assess the safety and effectiveness of a polyphenol-based remedial strategy. The final supplement represents a product of an appropriate technological process, conducted in accordance with the recommendations derived from the preceding research.
Collapse
Affiliation(s)
- Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Marko Miler
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Branka Šošić-Jurjević
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Branko Filipović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dragan Milenkovic
- Université Clermont Auvergne, INRAE, UNH, F-63000 Clermont-Ferrand, France
| | - Vladimir Jakovljević
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia; Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, Moscow, Russian Federation
| | - Verica Milošević
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
23
|
Canet-Pons J, Sen NE, Arsović A, Almaguer-Mederos LE, Halbach MV, Key J, Döring C, Kerksiek A, Picchiarelli G, Cassel R, René F, Dieterlé S, Fuchs NV, König R, Dupuis L, Lütjohann D, Gispert S, Auburger G. Atxn2-CAG100-KnockIn mouse spinal cord shows progressive TDP43 pathology associated with cholesterol biosynthesis suppression. Neurobiol Dis 2021; 152:105289. [PMID: 33577922 DOI: 10.1016/j.nbd.2021.105289] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/11/2020] [Accepted: 02/03/2021] [Indexed: 12/12/2022] Open
Abstract
Large polyglutamine expansions in Ataxin-2 (ATXN2) cause multi-system nervous atrophy in Spinocerebellar Ataxia type 2 (SCA2). Intermediate size expansions carry a risk for selective motor neuron degeneration, known as Amyotrophic Lateral Sclerosis (ALS). Conversely, the depletion of ATXN2 prevents disease progression in ALS. Although ATXN2 interacts directly with RNA, and in ALS pathogenesis there is a crucial role of RNA toxicity, the affected functional pathways remain ill defined. Here, we examined an authentic SCA2 mouse model with Atxn2-CAG100-KnockIn for a first definition of molecular mechanisms in spinal cord pathology. Neurophysiology of lower limbs detected sensory neuropathy rather than motor denervation. Triple immunofluorescence demonstrated cytosolic ATXN2 aggregates sequestrating TDP43 and TIA1 from the nucleus. In immunoblots, this was accompanied by elevated CASP3, RIPK1 and PQBP1 abundance. RT-qPCR showed increase of Grn, Tlr7 and Rnaset2 mRNA versus Eif5a2, Dcp2, Uhmk1 and Kif5a decrease. These SCA2 findings overlap well with known ALS features. Similar to other ataxias and dystonias, decreased mRNA levels for Unc80, Tacr1, Gnal, Ano3, Kcna2, Elovl5 and Cdr1 contrasted with Gpnmb increase. Preterminal stage tissue showed strongly activated microglia containing ATXN2 aggregates, with parallel astrogliosis. Global transcriptome profiles from stages of incipient motor deficit versus preterminal age identified molecules with progressive downregulation, where a cluster of cholesterol biosynthesis enzymes including Dhcr24, Msmo1, Idi1 and Hmgcs1 was prominent. Gas chromatography demonstrated a massive loss of crucial cholesterol precursor metabolites. Overall, the ATXN2 protein aggregation process affects diverse subcellular compartments, in particular stress granules, endoplasmic reticulum and receptor tyrosine kinase signaling. These findings identify new targets and potential biomarkers for neuroprotective therapies.
Collapse
Affiliation(s)
- Júlia Canet-Pons
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Nesli-Ece Sen
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany; Faculty of Biosciences, Goethe University, 60438 Frankfurt am Main, Germany
| | - Aleksandar Arsović
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Luis-Enrique Almaguer-Mederos
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany; Center for Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba
| | - Melanie V Halbach
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Jana Key
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany; Faculty of Biosciences, Goethe University, 60438 Frankfurt am Main, Germany
| | - Claudia Döring
- Dr. Senckenberg Institute of Pathology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Nordrhein-Westfalen, Germany
| | - Gina Picchiarelli
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Raphaelle Cassel
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Frédérique René
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Stéphane Dieterlé
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Nina V Fuchs
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Renate König
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Luc Dupuis
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Nordrhein-Westfalen, Germany
| | - Suzana Gispert
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Georg Auburger
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
24
|
Bosch-Queralt M, Cantuti-Castelvetri L, Damkou A, Schifferer M, Schlepckow K, Alexopoulos I, Lütjohann D, Klose C, Vaculčiaková L, Masuda T, Prinz M, Monroe KM, Di Paolo G, Lewcock JW, Haass C, Simons M. Diet-dependent regulation of TGFβ impairs reparative innate immune responses after demyelination. Nat Metab 2021; 3:211-227. [PMID: 33619376 PMCID: PMC7610359 DOI: 10.1038/s42255-021-00341-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023]
Abstract
Proregenerative responses are required for the restoration of nervous-system functionality in demyelinating diseases such as multiple sclerosis (MS). Yet, the limiting factors responsible for poor CNS repair are only partially understood. Here, we test the impact of a Western diet (WD) on phagocyte function in a mouse model of demyelinating injury that requires microglial innate immune function for a regenerative response to occur. We find that WD feeding triggers an ageing-related, dysfunctional metabolic response that is associated with impaired myelin-debris clearance in microglia, thereby impairing lesion recovery after demyelination. Mechanistically, we detect enhanced transforming growth factor beta (TGFβ) signalling, which suppresses the activation of the liver X receptor (LXR)-regulated genes involved in cholesterol efflux, thereby inhibiting phagocytic clearance of myelin and cholesterol. Blocking TGFβ or promoting triggering receptor expressed on myeloid cells 2 (TREM2) activity restores microglia responsiveness and myelin-debris clearance after demyelinating injury. Thus, we have identified a druggable microglial immune checkpoint mechanism regulating the microglial response to injury that promotes remyelination.
Collapse
Affiliation(s)
- Mar Bosch-Queralt
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Ludovico Cantuti-Castelvetri
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Ioannis Alexopoulos
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Dieter Lütjohann
- Institute for of Clinical Chemistry and Clinical Pharmacology, University of Hospital Bonn, Bonn, Germany
| | | | - Lenka Vaculčiaková
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | | | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
25
|
Jahn T, Clark C, Kerksiek A, Lewczuk P, Lütjohann D, Popp J. Cholesterol metabolites and plant sterols in cerebrospinal fluid are associated with Alzheimer's cerebral pathology and clinical disease progression. J Steroid Biochem Mol Biol 2021; 205:105785. [PMID: 33171206 DOI: 10.1016/j.jsbmb.2020.105785] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND PURPOSE Altered cholesterol metabolism is associated with increased risk of neurodegeneration and in particular with the development of Alzheimer's disease (AD). Here, we investigate whether non-cholesterol sterols and oxysterols in the central nervous system are associated with (i) the presence of cerebral AD pathology, (ii) distinct aspects of AD pathology, i.e. amyloid pathology, neuronal injury, and tau pathology, and (iii) cognitive decline over time. EXPERIMENTAL APPROACH One hundred forty-two elder subjects with normal cognition, mild cognitive impairment, or mild dementia participating in a cohort study on cognitive decline and AD were included. Clinical and neuropsychological assessments were performed at inclusion and repeated at follow-up visits at 18 and 36 months. Concentrations of cholesterol, non-cholesterol sterols, and cholesterol metabolites were measured in cerebrospinal fluid (CSF), along with CSF beta-amyloid (Aβ)1-42; Aβ1-42/Aβ1-40 ratio, total-tau (tau), and tau phosphorylated at threonine 181 (p-tau) as markers of amyloid pathology, neuronal injury and tau pathology, respectively. Cognitive decline was assessed by changes in Mini-Mental State Examination and Clinical Dementia Rating sum of boxes at follow-up visits. KEY RESULTS CSF 24S-hydroxycholesterol (24S-OHC) and the 24S-OHC/27-OHC ratio were higher in subjects with AD pathology. CSF desmosterol correlated with Aβ1-42 levels. The 24S-OHC levels, the 24S-OHC/27-OHC ratio and the plant sterols campesterol and sitosterol were associated with the tau and p-tau levels. Both plant sterol concentrations along with the 24S-OHC/27-OHC ratio at baseline predicted cognitive decline at follow-up visits. CONCLUSIONS AND IMPLICATIONS We show the importance of CSF levels of several non-cholesterol sterols and oxysterols to AD and core AD biomarkers. The plant sterols campesterol and sitosterol appear to be involved in tau pathology and neurodegeneration. CSF desmosterol level indicates CNS cholesterol synthesis and might be of relevance for clinical disease severity. Therefore these non-cholesterol sterols may represent intervention targets to slow down disease progression.
Collapse
Affiliation(s)
- Tabea Jahn
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Christopher Clark
- Institute for Regenerative Medicine, University of Zürich, Zürich, Switzerland
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, University Clinic Erlangen, and Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Department of Neurodegeneration Diagnostics and Department of Biochemical Diagnostics, University Hospital of Bialystok, Bialystok, Poland
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.
| | - Julius Popp
- Institute for Regenerative Medicine, University of Zürich, Zürich, Switzerland; Department of Psychiatry, University Hospital of Lausanne, Switzerland; Department of Geriatric Psychiatry, University Hospital of Psychiatry Zurich, Zürich, Switzerland.
| |
Collapse
|
26
|
Lütjohann D, Stellaard F, Kerksiek A, Lötsch J, Oertel BG. Serum 4β-hydroxycholesterol increases during fluconazole treatment. Eur J Clin Pharmacol 2020; 77:659-669. [PMID: 33201347 PMCID: PMC8032583 DOI: 10.1007/s00228-020-03041-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/09/2020] [Indexed: 11/02/2022]
Abstract
PURPOSE The antifungal drugs ketoconazole and itraconazole reduce serum concentrations of 4β-hydroxycholesterol, which is a validated marker for hepatic cytochrome P450 (CYP) 3A4 activity. We tested the effect of another antifungal triazole agent, fluconazole, on serum concentrations of different sterols and oxysterols within the cholesterol metabolism to see if this inhibitory reaction is a general side effect of azole antifungal agents. METHODS In a prospective, double-blind, placebo-controlled, two-way crossover design, we studied 17 healthy subjects (nine men, eight women) who received 400 mg fluconazole or placebo daily for 8 days. On day 1 before treatment and on day 8 after the last dose, fasting blood samples were collected. Serum cholesterol precursors and oxysterols were measured by gas chromatography-mass spectrometry-selected ion monitoring and expressed as the ratio to cholesterol (R_sterol). RESULTS Under fluconazole treatment, serum R_lanosterol and R_24,25-dihydrolanosterol increased significantly without affecting serum cholesterol or metabolic downstream markers of hepatic cholesterol synthesis. Serum R_4β-, R_24S-, and R_27-hydroxycholesterol increased significantly. CONCLUSION Fluconazole inhibits the 14α-demethylation of lanosterol and 24,25-dihydrolanosterol, regulated by CYP51A1, without reduction of total cholesterol synthesis. The increased serum level of R_4β-hydroxycholesterol under fluconazole treatment is in contrast to the reductions observed under ketoconazole and itraconazole treatments. The question, whether this increase is caused by induction of CYP3A4 or by inhibition of the catabolism of 4β-hydroxycholesterol, must be answered by mechanistic in vitro and in vivo studies comparing effects of various azole antifungal agents on hepatic CYP3A4 activity.
Collapse
Affiliation(s)
- Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Frans Stellaard
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor Stern Kai 7, 60590, Frankfurt, Germany
| | - Bruno G Oertel
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor Stern Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
27
|
Zeitz JO, Ehbrecht T, Fleischmann A, Most E, Gessner DK, Friedrichs S, Sparenberg M, Failing K, Whelan R, Lütjohann D, Eder K. Effect of DL-Methionine Supplementation on Tissue and Plasma Antioxidant Status and Concentrations of Oxidation Products of Cholesterol and Phytosterols in Heat-Processed Thigh Muscle of Broilers. Animals (Basel) 2020; 10:E2050. [PMID: 33167600 PMCID: PMC7694460 DOI: 10.3390/ani10112050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
In this study, the hypothesis that supplementation with methionine (Met) as DL-Met (DLM) in excess of the National Research Council (NRC) recommendations improves the antioxidant system in broilers was investigated. Day-old male Cobb-500 broilers (n = 72) were divided into three groups which were fed a control diet or diets supplemented with two levels of DLM in which the concentrations of Met + Cys exceeded the recommendations of NRC by 15-20% (group DLM 1) or 30-40% (group DLM 2), respectively. The three groups of broilers did not show differences in body weight gains, feed intake, and feed conversion ratio. However, broilers of groups DLM 1 and DLM 2 had higher concentrations of glutathione (GSH) in liver and thigh muscle and lower concentrations of cholesterol oxidation products (COPs) in heat-processed thigh muscle than broilers of the control group. Concentrations of several oxidation products of phytosterols in heat-processed thigh muscle were also reduced in groups DLM 1 and DLM 2; however, the concentration of total oxidation products of phytosterols was not different between the three groups. The study shows that DLM supplementation improved the antioxidant status due to an increased formation of GSH and reduced the formation of COPs during heat-processing in thigh muscle.
Collapse
Affiliation(s)
- Johanna O. Zeitz
- Institute of Animal Nutrition and Nutritional Physiology, Justus Liebig University, D-35392 Giessen, Germany; (J.O.Z.); (T.E.); (A.F.); (E.M.); (D.K.G.)
| | - Tamara Ehbrecht
- Institute of Animal Nutrition and Nutritional Physiology, Justus Liebig University, D-35392 Giessen, Germany; (J.O.Z.); (T.E.); (A.F.); (E.M.); (D.K.G.)
| | - Anne Fleischmann
- Institute of Animal Nutrition and Nutritional Physiology, Justus Liebig University, D-35392 Giessen, Germany; (J.O.Z.); (T.E.); (A.F.); (E.M.); (D.K.G.)
| | - Erika Most
- Institute of Animal Nutrition and Nutritional Physiology, Justus Liebig University, D-35392 Giessen, Germany; (J.O.Z.); (T.E.); (A.F.); (E.M.); (D.K.G.)
| | - Denise K. Gessner
- Institute of Animal Nutrition and Nutritional Physiology, Justus Liebig University, D-35392 Giessen, Germany; (J.O.Z.); (T.E.); (A.F.); (E.M.); (D.K.G.)
| | - Silvia Friedrichs
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, D-53127 Bonn, Germany; (S.F.); (D.L.)
| | - Marion Sparenberg
- Unit of Biomathematics and Data Processing, Faculty of Veterinary Medicine, Justus Liebig University, D-35392 Giessen, Germany; (M.S.); (K.F.)
| | - Klaus Failing
- Unit of Biomathematics and Data Processing, Faculty of Veterinary Medicine, Justus Liebig University, D-35392 Giessen, Germany; (M.S.); (K.F.)
| | - Rose Whelan
- Evonik Operations GmbH, D-63457 Hanau-Wolfgang, Germany;
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, D-53127 Bonn, Germany; (S.F.); (D.L.)
| | - Klaus Eder
- Institute of Animal Nutrition and Nutritional Physiology, Justus Liebig University, D-35392 Giessen, Germany; (J.O.Z.); (T.E.); (A.F.); (E.M.); (D.K.G.)
| |
Collapse
|
28
|
Alipour MR, Karimi-Sales E. Molecular mechanisms of protective roles of isoflavones against chemicals-induced liver injuries. Chem Biol Interact 2020; 329:109213. [DOI: 10.1016/j.cbi.2020.109213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/27/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
|
29
|
Gutierrez E, Lütjohann D, Kerksiek A, Fabiano M, Oikawa N, Kuerschner L, Thiele C, Walter J. Importance of γ-secretase in the regulation of liver X receptor and cellular lipid metabolism. Life Sci Alliance 2020; 3:3/6/e201900521. [PMID: 32354700 PMCID: PMC7195048 DOI: 10.26508/lsa.201900521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Inhibition of the Alzheimer associated γ-secretase impairs the regulation of cellular lipid droplet homeostasis. Presenilins (PS) are the catalytic components of γ-secretase complexes that mediate intramembrane proteolysis. Mutations in the PS genes are a major cause of familial early-onset Alzheimer disease and affect the cleavage of the amyloid precursor protein, thereby altering the production of the amyloid β-peptide. However, multiple additional protein substrates have been identified, suggesting pleiotropic functions of γ-secretase. Here, we demonstrate that inhibition of γ-secretase causes dysregulation of cellular lipid homeostasis, including up-regulation of liver X receptors, and complex changes in the cellular lipid composition. Genetic and pharmacological inhibition of γsecretase leads to strong accumulation of cytoplasmic lipid droplets, associated with increased levels of acylglycerols, but lowered cholesteryl esters. Furthermore, accumulation of lipid droplets was augmented by increasing levels of amyloid precursor protein C-terminal fragments, indicating a critical involvement of this γ-secretase substrate. Together, these data provide a mechanism that functionally connects γ-secretase activity to cellular lipid metabolism. These effects were also observed in human astrocytic cells, indicating an important function of γ-secretase in cells critical for lipid homeostasis in the brain.
Collapse
Affiliation(s)
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Marietta Fabiano
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Naoto Oikawa
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Lars Kuerschner
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Christoph Thiele
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Jochen Walter
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
30
|
Ajdžanović V, Miler M, Živanović J, Filipović B, Šošić-Jurjević B, Popovska-Perčinić F, Milošević V. The adrenal cortex after estradiol or daidzein application in a rat model of the andropause: Structural and hormonal study. Ann Anat 2020; 230:151487. [PMID: 32120001 DOI: 10.1016/j.aanat.2020.151487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION AND AIM Daidzein application may represent an effective and less harmful alternative to indicated, classical estrogenization of ageing men. The aim of this study was to perform structural and hormonal analysis of the adrenal cortex, after estradiol or daidzein supplementation in a rat model of the andropause. MATERIAL AND METHODS Middle-aged Wistar rats were divided into sham operated (SO; n = 8), orchidectomized (Orx; n = 8), estradiol treated orchidectomized (Orx + E; n = 8) and daidzein treated orchidectomized (Orx + D; n = 8) groups. Estradiol (0.625 mg/kg b.m./day) or daidzein (30 mg/kg b.m./day) were administered subcutaneously for three weeks, while the SO and Orx groups received the vehicle alone. Set objectives were achieved using stereology, histochemistry/immunohistochemistry, immunoassays and ultrastructural analysis. RESULTS Both estradiol and daidzein treatment significantly increased volumes of the zona glomerulosa cell and nuclei, but decreased circulating aldosterone levels. Estradiol markedly increased volumes of the zona fasciculata cell and nuclei in parallel with significant decrease of the adrenal tissue level of corticosterone, while daidzein significantly decreased both the adrenal and circulating levels of corticosterone. Serum DHEA level and volumes of the zona reticularis cell and nuclei significantly increased upon estradiol treatment, whereas daidzein even stronger increased the circulating level of DHEA. Shunting of the corticosteroidogenesis pathways towards adrenal androgens production, after the treatments, corresponded to the ultrastructural findings and zonal capillary network rearrangements. CONCLUSIONS Given the coherence of its effects and relative safety, daidzein could be the remedy of choice for the treatment of ageing-caused androgen deprivation and the hypothalamo-pituitary-adrenal axis hyperfunction/related metabolic issues in males.
Collapse
Affiliation(s)
- Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Marko Miler
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jasmina Živanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Branko Filipović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Branka Šošić-Jurjević
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | | | - Verica Milošević
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
31
|
Yalcinkaya M, Kerksiek A, Gebert K, Annema W, Sibler R, Radosavljevic S, Lütjohann D, Rohrer L, von Eckardstein A. HDL inhibits endoplasmic reticulum stress-induced apoptosis of pancreatic β-cells in vitro by activation of Smoothened. J Lipid Res 2020; 61:492-504. [PMID: 31907205 DOI: 10.1194/jlr.ra119000509] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/18/2019] [Indexed: 01/20/2023] Open
Abstract
Loss of pancreatic β-cell mass and function as a result of sustained ER stress is a core step in the pathogenesis of diabetes mellitus type 2. The complex control of β-cells and insulin production involves hedgehog (Hh) signaling pathways as well as cholesterol-mediated effects. In fact, data from studies in humans and animal models suggest that HDL protects against the development of diabetes through inhibition of ER stress and β-cell apoptosis. We investigated the mechanism by which HDL inhibits ER stress and apoptosis induced by thapsigargin, a sarco/ER Ca2+-ATPase inhibitor, in β-cells of a rat insulinoma cell line, INS1e. We further explored effects on the Hh signaling receptor Smoothened (SMO) with pharmacologic agonists and inhibitors. Interference with sterol synthesis or efflux enhanced β-cell apoptosis and abrogated the anti-apoptotic activity of HDL. During ER stress, HDL facilitated the efflux of specific oxysterols, including 24-hydroxycholesterol (OHC). Supplementation of reconstituted HDL with 24-OHC enhanced and, in cells lacking ABCG1 or the 24-OHC synthesizing enzyme CYP46A1, restored the protective activity of HDL. Inhibition of SMO countered the beneficial effects of HDL and also LDL, and SMO agonists decreased β-cell apoptosis in the absence of ABCG1 or CYP46A1. The translocation of the SMO-activated transcription factor glioma-associated oncogene GLI-1 was inhibited by ER stress but restored by both HDL and 24-OHC. In conclusion, the protective effect of HDL to counter ER stress and β-cell death involves the transport, generation, and mobilization of oxysterols for activation of the Hh signaling receptor SMO.
Collapse
Affiliation(s)
- Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Katrin Gebert
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Wijtske Annema
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Rahel Sibler
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Silvija Radosavljevic
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Šošić-Jurjević B, Ajdžanović V, Filipović B, Severs W, Milošević V. Thyroid Mediation of the Isoflavone Effects on Osteoporotic Bone: The Endocrine Interference With a Beneficial Outcome. Front Endocrinol (Lausanne) 2019; 10:688. [PMID: 31681166 PMCID: PMC6798150 DOI: 10.3389/fendo.2019.00688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/23/2019] [Indexed: 01/11/2023] Open
Affiliation(s)
- Branka Šošić-Jurjević
- Department of Cytology, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Branko Filipović
- Department of Cytology, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Walter Severs
- College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Verica Milošević
- Department of Cytology, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| |
Collapse
|