1
|
Zeng X, Wang Y, Farias K, Rappa A, Darko C, Sauve A, Huang Q, Alonso LC, Yang Y. NRH, a potent NAD + enhancer, improves glucose homeostasis and lipid metabolism in diet-induced obese mice through an active adenosine kinase pathway. Metabolism 2025; 164:156110. [PMID: 39710001 PMCID: PMC11788054 DOI: 10.1016/j.metabol.2024.156110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
AIMS NAD+ deficiency underlies obesity-induced metabolic disturbances. This study evaluated dihydronicotinamide riboside (NRH), a potent NAD+ enhancer, in lean and obese mice and explored whether NRH operates through a unique mechanism involving adenosine kinase (ADK), an enzyme critical for NRH-driven NAD+ synthesis. METHODS Pharmacokinetic and pharmacodynamic analyses were performed following a single 250 mg/kg intraperitoneal injection of NRH in healthy mice. In long-term studies, lean and high-fat diet-induced obese mice were treated with 250 mg/kg NRH thrice weekly for 7 weeks. Blood NAD+ levels, body composition, energy expenditure, and glucose and lipid metabolism were monitored. To test ADK's role, the ADK inhibitor ABT702 was co-administered with NRH in obese mice. RESULTS NRH entered tissues unassisted and was rapidly metabolized for NAD+ biosynthesis, while ADK inhibition blocked its phosphorylation, leading to NRH accumulation in all examined tissues and possible release back into circulation. The 7-week NRH administration was well-tolerated in both lean and obese mice. In obese mice, NRH improved glucose homeostasis by boosting insulin secretion, enhancing muscle insulin signaling, and reducing hepatic gluconeogenesis. It also lowered fat mass, decreased serum lipids, and improved white adipose function. These benefits were linked to elevated tissue NAD+ levels, enhanced Sirtuin activities, and increased mitochondrial antioxidant defenses. ADK inhibition abolished these effects, confirming that NRH's direct entry into tissues and subsequent phosphorylation is essential for its full benefits. CONCLUSION This study establishes NRH as a promising therapeutic agent for obesity-induced metabolic dysfunction, correcting glucose intolerance and hyperlipidemia through ADK-dependent NAD+ enhancement.
Collapse
Affiliation(s)
- Xinliu Zeng
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongjie Wang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Department of Animal Sciences, North Carolina A&T State University, Greensboro, NC 27411, United States of America
| | - Karina Farias
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Andrew Rappa
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Christine Darko
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Anthony Sauve
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Qingxia Huang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Laura C Alonso
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Yue Yang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| |
Collapse
|
2
|
Pecci F, Cognigni V, Giudice GC, Paoloni F, Cantini L, Saini KS, Abushukair HM, Naqash AR, Cortellini A, Mazzaschi G, Alia S, Membrino V, Araldi E, Tiseo M, Buti S, Vignini A, Berardi R. Unraveling the link between cholesterol and immune system in cancer: From biological mechanistic insights to clinical evidence. A narrative review. Crit Rev Oncol Hematol 2025; 209:104654. [PMID: 39923921 DOI: 10.1016/j.critrevonc.2025.104654] [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: 11/26/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
Cholesterol and its metabolism seem to be involved not only in cancer progression but also in immune cells activity. In this comprehensive review, we summarize preclinical, translational, and clinical evidence regarding the crucial role of cholesterol and its metabolism in regulating the immune response against cancer cells, shedding light on the multifaceted mechanisms by which cholesterol influences immune cell function and anti-tumor immunity. By synthesizing findings from preclinical studies, we have elucidated the impact of cholesterol on immune cell activation, differentiation, and effector functions. These investigations have revealed that cholesterol metabolism plays a pivotal role in shaping the immune response, with alterations in cholesterol levels directly impacting immune cell behavior and anti-tumor activity. All the steps related to cholesterol metabolism, including its de-novo synthesis, its influx and efflux mechanisms, as well as its metabolites, have a distinct impact on immune cells function and activity, which, if altered, might influence tumor progression. In addition, we have reviewed clinical studies investigating the role of circulating cholesterol on outcomes of patients with advanced tumors treated with immune checkpoint inhibitors, highlighting again in a clinical scenario the correlation between cholesterol and the immune system. Overall, our review emphasizes the importance of cholesterol and its metabolism in orchestrating the immune response against cancer cells. Herein we have provided a comprehensive overview of this emerging field by illustrating the intricate interplay between cholesterol and immune system.
Collapse
Affiliation(s)
- Federica Pecci
- Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy.
| | - Valeria Cognigni
- Department of Medical Oncology, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| | - Giulia Claire Giudice
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Francesco Paoloni
- Department of Medical Oncology, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| | | | - Kamal S Saini
- Fortrea, Inc., Durham, NC, USA; Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Hassan Mohammed Abushukair
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, USA; Division of Oncology Sciences, University of Oklahoma Health Sciences Center, USA
| | - Abdul Rafeh Naqash
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, USA; Division of Oncology Sciences, University of Oklahoma Health Sciences Center, USA; Medical Oncology/TSET Phase 1 Program, Stephenson Cancer Center, The University of Oklahoma, Oklahoma City, OK, USA
| | - Alessio Cortellini
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy; Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Giulia Mazzaschi
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Sonila Alia
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Valentina Membrino
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Elisa Araldi
- Systems Medicine Laboratory, Department of Medicine and Surgery (DiMeC), Università degli Studi di Parma, Parma, Italy; Preventive Cardiology and Preventive Medicine, University Medical Center Of The Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Germany
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Sebastiano Buti
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Arianna Vignini
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy; Research Center of Health Education and Health Promotion, Università Politecnica delle Marche, Ancona, Italy
| | - Rossana Berardi
- Department of Medical Oncology, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| |
Collapse
|
3
|
Hidrobo MS, Höring M, Brunner S, Liebisch G, Schweizer S, Klingenspor M, Schreiber R, Zechner R, Burkhardt R, Ecker J. Cold-induced phosphatidylethanolamine synthesis in liver and brown adipose tissue of mice. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159562. [PMID: 39214167 DOI: 10.1016/j.bbalip.2024.159562] [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: 04/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Increasing energy expenditure in brown adipose (BAT) tissue by cold-induced lipolysis is discussed as a potential strategy to counteract imbalanced lipid homeostasis caused through unhealthy lifestyle and cardiometabolic disease. Yet, it is largely unclear how liberated fatty acids (FA) are metabolized. We investigated the liver and BAT lipidome of mice housed for 1 week at thermoneutrality, 23 °C and 4 °C using quantitative mass spectrometry-based lipidomics. Housing at temperatures below thermoneutrality triggered the generation of phosphatidylethanolamine (PE) in both tissues. Particularly, the concentrations of PE containing polyunsaturated fatty acids (PUFA) in their acyl chains like PE 18:0_20:4 were increased at cold. Investigation of the plasma's FA profile using gas chromatography coupled to mass spectrometry revealed a negative correlation of PUFA with unsaturated PE in liver and BAT indicating a flux of FA from the circulation into these tissues. Beta-adrenergic stimulation elevated intracellular levels of PE 38:4 and PE 40:6 in beige wildtype adipocytes, but not in adipose triglyceride lipase (ATGL)-deficient cells. These results imply an induction of PE synthesis in liver, BAT and thermogenic adipocytes after activation of the beta-adrenergic signaling cascade.
Collapse
Affiliation(s)
- Maria Soledad Hidrobo
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Sarah Brunner
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Sabine Schweizer
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/2, 8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/2, 8010 Graz, Austria
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Josef Ecker
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany.
| |
Collapse
|
4
|
Höring M, Brunner S, Scheiber J, Honecker J, Liebisch G, Seeliger C, Schinhammer L, Claussnitzer M, Burkhardt R, Hauner H, Ecker J. Sex-specific response of the human plasma lipidome to short-term cold exposure. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159567. [PMID: 39366508 DOI: 10.1016/j.bbalip.2024.159567] [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: 05/24/2024] [Revised: 09/05/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Cold-induced lipolysis is widely studied as a potential therapeutic strategy to combat metabolic disease, but its effect on lipid homeostasis in humans remains largely unclear. Blood plasma comprises an enormous repertoire in lipids allowing insights into whole body lipid homeostasis. So far, reported results originate from studies carried out with small numbers of male participants. Here, the blood plasma's lipidome of 78 male and 93 female volunteers, who were exposed to cold below the shivering threshold for 2 h, was quantified by comprehensive lipidomics using high-resolution mass spectrometry. Short-term cold exposure increased the concentrations in 147 of 177 quantified circulating lipids and the response of the plasma's lipidome was sex-specific. In particular, the amounts of generated glycerophospholipid and sphingolipid species differed between the sexes. In women, the BMI could be related with the lipidome's response. A logistic regression model predicted with high sensitivity and specificity whether plasma samples were from male or female subjects based on the cold-induced response of phosphatidylcholine (PC), lysophosphatidylcholine (LPC), and sphingomyelin (SM) species. In summary, cold exposure promotes lipid synthesis by supplying fatty acids generated after lipolysis for all lipid classes. The plasma lipidome, i.e. PC, LPC and SM, shows a sex-specific response, indicating a different regulation of its metabolism in men and women. This supports the need for sex-specific research and avoidance of sex bias in clinical trials.
Collapse
Affiliation(s)
- Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Sarah Brunner
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | | | - Julius Honecker
- Institute of Nutritional Medicine, Else Kröner Fresenius Centre for Nutritional Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Claudine Seeliger
- Institute of Nutritional Medicine, Else Kröner Fresenius Centre for Nutritional Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Laura Schinhammer
- Institute of Nutritional Medicine, Else Kröner Fresenius Centre for Nutritional Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Melina Claussnitzer
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Hans Hauner
- Institute of Nutritional Medicine, Else Kröner Fresenius Centre for Nutritional Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany.
| | - Josef Ecker
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
5
|
Cox LA, Puppala S, Chan J, Riojas AM, Lange KJ, Birnbaum S, Dick EJ, Comuzzie AG, Nijland MJ, Li C, Nathanielsz PW, Olivier M. Maternal under-nutrition during pregnancy alters the molecular response to over-nutrition in multiple organs and tissues in nonhuman primate juvenile offspring. J Dev Orig Health Dis 2024; 15:e27. [PMID: 39506415 DOI: 10.1017/s2040174424000163] [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] [Indexed: 11/08/2024]
Abstract
Previous studies in rodents suggest that mismatch between fetal and postnatal nutrition predisposes individuals to metabolic diseases. We hypothesized that in nonhuman primates (NHP), fetal programming of maternal undernutrition (MUN) persists postnatally with a dietary mismatch altering metabolic molecular systems that precede standard clinical measures. We used unbiased molecular approaches to examine response to a high fat, high-carbohydrate diet plus sugar drink (HFCS) challenge in NHP juvenile offspring of MUN pregnancies compared with controls (CON). Pregnant baboons were fed ad libitum (CON) or 30% calorie reduction from 0.16 gestation through lactation; weaned offspring were fed chow ad libitum. MUN offspring were growth restricted at birth. Liver, omental fat, and skeletal muscle gene expression, and liver glycogen, muscle mitochondria, and fat cell size were quantified. Before challenge, MUN offspring had lower body mass index (BMI) and liver glycogen, and consumed more sugar drink than CON. After HFCS challenge, MUN and CON BMIs were similar. Molecular analyses showed HFCS response differences between CON and MUN for muscle and liver, including hepatic splicing and unfolded protein response. Altered liver signaling pathways and glycogen content between MUN and CON at baseline indicate in utero programming persists in MUN juveniles. MUN catchup growth during consumption of HFCS suggests increased risk of obesity, diabetes, and cardiovascular disease. Greater sugar drink consumption in MUN demonstrates altered appetitive drive due to programming. Differences in blood leptin, liver glycogen, and tissue-specific molecular response to HFCS suggest MUN significantly impacts juvenile offspring ability to manage an energy rich diet.
Collapse
Affiliation(s)
- Laura A Cox
- Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Sobha Puppala
- Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jeannie Chan
- Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Angelica M Riojas
- Department of Radiology, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Shifra Birnbaum
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | | | - Cun Li
- Department of Animal Sciences, University of Wyoming, Laramie, WY, USA
| | - Peter W Nathanielsz
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Animal Sciences, University of Wyoming, Laramie, WY, USA
| | - Michael Olivier
- Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
6
|
Weiss U, Mungo E, Haß M, Benning D, Gurke R, Hahnefeld L, Dorochow E, Schlaudraff J, Schmid T, Kuntschar S, Meyer S, Medert R, Freichel M, Geisslinger G, Niederberger E. Knock-Out of IKKepsilon Ameliorates Atherosclerosis and Fatty Liver Disease by Alterations of Lipid Metabolism in the PCSK9 Model in Mice. Int J Mol Sci 2024; 25:10721. [PMID: 39409049 PMCID: PMC11476531 DOI: 10.3390/ijms251910721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
The inhibitor-kappaB kinase epsilon (IKKε) represents a non-canonical IκB kinase that modulates NF-κB activity and interferon I responses. Inhibition of this pathway has been linked with atherosclerosis and metabolic dysfunction-associated steatotic liver disease (MASLD), yet the results are contradictory. In this study, we employed a combined model of hepatic PCSK9D377Y overexpression and a high-fat diet for 16 weeks to induce atherosclerosis and liver steatosis. The development of atherosclerotic plaques, serum lipid concentrations, and lipid metabolism in the liver and adipose tissue were compared between wild-type and IKKε knock-out mice. The formation and progression of plaques were markedly reduced in IKKε knockout mice, accompanied by reduced serum cholesterol levels, fat deposition, and macrophage infiltration within the plaque. Additionally, the development of a fatty liver was diminished in these mice, which may be attributed to decreased levels of multiple lipid species, particularly monounsaturated fatty acids, triglycerides, and ceramides in the serum. The modulation of several proteins within the liver and adipose tissue suggests that de novo lipogenesis and the inflammatory response are suppressed as a consequence of IKKε inhibition. In conclusion, our data suggest that the knockout of IKKε is involved in mechanisms of both atherosclerosis and MASLD. Inhibition of this pathway may therefore represent a novel approach to the treatment of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ulrike Weiss
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Eleonora Mungo
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Michelle Haß
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Denis Benning
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Robert Gurke
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Lisa Hahnefeld
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Erika Dorochow
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
| | - Jessica Schlaudraff
- Goethe University Frankfurt, Faculty of Medicine, Institute of Neuroanatomy, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany;
| | - Tobias Schmid
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Silvia Kuntschar
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Sofie Meyer
- Goethe University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (T.S.); (S.K.); (S.M.)
| | - Rebekka Medert
- Institute of Pharmacology, Ruprechts-Karl University Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (R.M.); (M.F.)
| | - Marc Freichel
- Institute of Pharmacology, Ruprechts-Karl University Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (R.M.); (M.F.)
| | - Gerd Geisslinger
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Ellen Niederberger
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (U.W.); (E.M.); (M.H.); (D.B.); (R.G.); (L.H.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
7
|
Dixon ED, Claudel T, Nardo AD, Riva A, Fuchs CD, Mlitz V, Busslinger G, Scharnagl H, Stojakovic T, Senéca J, Hinteregger H, Grabner GF, Kratky D, Verkade H, Zimmermann R, Haemmerle G, Trauner M. Inhibition of ATGL alleviates MASH via impaired PPARα signalling that favours hydrophilic bile acid composition in mice. J Hepatol 2024:S0168-8278(24)02577-7. [PMID: 39357546 DOI: 10.1016/j.jhep.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND & AIMS Adipose triglyceride lipase (ATGL) is an attractive therapeutic target in insulin resistance and metabolic dysfunction-associated steatotic liver disease (MASLD). This study investigated the effects of pharmacological ATGL inhibition on the development of metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis in mice. METHODS Streptozotocin-injected male mice were fed a high-fat diet to induce MASH. Mice receiving the ATGL inhibitor atglistatin (ATGLi) were compared to controls using liver histology, lipidomics, metabolomics, 16s rRNA, and RNA sequencing. Human ileal organoids, HepG2 cells, and Caco2 cells treated with the human ATGL inhibitor NG-497, HepG2 ATGL knockdown cells, gel-shift, and luciferase assays were analysed for mechanistic insights. We validated the benefits of ATGLi on steatohepatitis and fibrosis in a low-methionine choline-deficient mouse model. RESULTS ATGLi improved serum liver enzymes, hepatic lipid content, and histological liver injury. Mechanistically, ATGLi attenuated PPARα signalling, favouring hydrophilic bile acid (BA) synthesis with increased Cyp7a1, Cyp27a1, Cyp2c70, and reduced Cyp8b1 expression. Additionally, reduced intestinal Cd36 and Abca1, along with increased Abcg5 expression, were consistent with reduced levels of hepatic triacylglycerol species containing polyunsaturated fatty acids, like linoleic acid, as well as reduced cholesterol levels in the liver and plasma. Similar changes in gene expression associated with PPARα signalling and intestinal lipid transport were observed in ileal organoids treated with NG-497. Furthermore, HepG2 ATGL knockdown cells revealed reduced expression of PPARα target genes and upregulation of genes involved in hydrophilic BA synthesis, consistent with reduced PPARα binding and luciferase activity in the presence of the ATGL inhibitors. CONCLUSIONS Inhibition of ATGL attenuates PPARα signalling, translating into hydrophilic BA composition, interfering with dietary lipid absorption, and improving metabolic disturbances. Validation with NG-497 opens a new therapeutic perspective for MASLD. IMPACT AND IMPLICATIONS Despite the recent approval of drugs novel mechanistic insights and pathophysiology-oriented therapeutic options for MASLD (metabolic dysfunction-associated steatotic liver disease) are still urgently needed. Herein, we show that pharmacological inhibition of ATGL, the key enzyme in lipid hydrolysis, using atglistatin (ATGLi), improves MASH (metabolic dysfunction-associated steatohepatitis), fibrosis, and key features of metabolic dysfunction in mouse models of MASH and liver fibrosis. Mechanistically, we demonstrated that attenuation of PPARα signalling in the liver and gut favours hydrophilic bile acid composition, ultimately interfering with dietary lipid absorption. One of the drawbacks of ATGLi is its lack of efficacy against human ATGL, thus limiting its clinical applicability. Against this backdrop, we could show that ATGL inhibition using the human inhibitor NG-497 in human primary ileum-derived organoids, Caco2 cells, and HepG2 cells translated into therapeutic mechanisms similar to ATGLi. Collectively, these findings reveal a possible new avenue for MASLD treatment.
Collapse
Affiliation(s)
- Emmanuel Dauda Dixon
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Alexander Daniel Nardo
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Alessandra Riva
- Chair of Nutrition and Immunology, School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Claudia Daniela Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Veronika Mlitz
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Georg Busslinger
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria; Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Tatjana Stojakovic
- Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, Austria
| | - Joana Senéca
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria; Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Helga Hinteregger
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Gernot F Grabner
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Dagmar Kratky
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Henkjan Verkade
- Department of Paediatrics, University Medical Centre Groningen, Groningen, Netherlands
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Zhang L, Wang X, Chen XW. The biogenesis and transport of triglyceride-rich lipoproteins. Trends Endocrinol Metab 2024:S1043-2760(24)00196-6. [PMID: 39164120 DOI: 10.1016/j.tem.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Triglyceride-rich lipoproteins (TRLs) play essential roles in human health and disease by transporting bulk lipids into the circulation. This review summarizes the fundamental mechanisms and diverse factors governing lipoprotein production, secretion, and regulation. Emphasizing the broader implications for human health, we outline the intricate landscape of lipoprotein research and highlight the potential coordination between the biogenesis and transport of TRLs in physiology, particularly the unexpected coupling of metabolic enzymes and transport machineries. Challenges and opportunities in lipoprotein biology with respect to inherited diseases and viral infections are also discussed. Further characterization of the biogenesis and transport of TRLs will advance both basic research in lipid biology and translational medicine for metabolic diseases.
Collapse
Affiliation(s)
- Linqi Zhang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China; Peking University (PKU)-Tsinghua University (THU) Joint Center for Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
9
|
Danner R, Prochniak LM, Pereckas M, Rouse JR, Wahhab A, Hackner LG, Lochhead RB. Identification of Major Histocompatibility Complex Class II Epitopes From Lyme Autoantigen Apolipoprotein B-100 and Borrelia burgdorferi Mcp4 in Murine Lyme Arthritis. J Infect Dis 2024; 230:S27-S39. [PMID: 39140726 PMCID: PMC11322890 DOI: 10.1093/infdis/jiae324] [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] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND During infection with the Lyme arthritis (LA) pathogen Borrelia burgdorferi, T-cell responses to both host and pathogen are dysregulated, resulting in chronic infection and frequent development of autoimmunity. METHODS To assess CD4+ T-cell epitopes presented during development of LA, we used an unbiased, immunopeptidomics approach to characterize the major histocompatibility complex (MHC) class II immunopeptidome in B burgdorferi-infected C57BL/6 (B6) mice, which develop mild, self-limiting LA, and infected B6 Il10-/- mice, which develop severe, persistent LA at 0, 4, and 16 weeks postinfection (22-23 mice per group). RESULTS Peptides derived from proteins involved in adaptive T- and B-cell responses and cholesterol metabolism, including human Lyme autoantigen apolipoprotein B-100 (apoB-100), were enriched in infected Il10-/- mice; whereas peptides derived from proteins involved in neutrophil extracellular net formation were enriched in infected B6 mice. Presentation of apoB-100 peptides showed evidence of epitope expansion during infection. Of several identified B burgdorferi peptides, only 1, a methyl-accepting chemotaxis protein peptide Mcp4442-462, was immunogenic. CONCLUSIONS ApoB-100, a human Lyme autoantigen, undergoes marked epitope expansion during LA development. The paucity of immunogenic B burgdorferi epitopes supports previous findings suggesting CD4+ T-cell responses are suppressed in murine LA.
Collapse
Affiliation(s)
- Rebecca Danner
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lauren M Prochniak
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michaela Pereckas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Joseph R Rouse
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Amanda Wahhab
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lauren G Hackner
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Robert B Lochhead
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
10
|
Amarsi R, Furse S, Cleaton MAM, Maurel S, Mitchell AL, Ferguson-Smith AC, Cenac N, Williamson C, Koulman A, Charalambous M. A co-ordinated transcriptional programme in the maternal liver supplies long chain polyunsaturated fatty acids to the conceptus using phospholipids. Nat Commun 2024; 15:6767. [PMID: 39117683 PMCID: PMC11310303 DOI: 10.1038/s41467-024-51089-z] [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: 07/11/2023] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
The long and very long chain polyunsaturated fatty acids (LC-PUFAs) are preferentially transported by the mother to the fetus. Failure to supply LC-PUFAs is strongly linked with stillbirth, fetal growth restriction, and impaired neurodevelopmental outcomes. However, dietary supplementation during pregnancy is unable to simply reverse these outcomes, suggesting imperfectly understood interactions between dietary fatty acid intake and the molecular mechanisms of maternal supply. Here we employ a comprehensive approach combining untargeted and targeted lipidomics with transcriptional profiling of maternal and fetal tissues in mouse pregnancy. Comparison of wild-type mice with genetic models of impaired lipid metabolism allows us to describe maternal hepatic adaptations required to provide LC-PUFAs to the developing fetus. A late pregnancy-specific, selective activation of the Liver X Receptor signalling pathway dramatically increases maternal supply of LC-PUFAs within circulating phospholipids. Crucially, genetic ablation of this pathway in the mother reduces LC-PUFA accumulation by the fetus, specifically of docosahexaenoic acid (DHA), a critical nutrient for brain development.
Collapse
Affiliation(s)
- Risha Amarsi
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, SE19RT, UK
- Pregnancy Physiology Laboratory, Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK
| | - Samuel Furse
- Biological chemistry group, Jodrell laboratory, Royal Botanic Gardens Kew, Kew Road, Richmond, Surrey, TW9 3DS, UK
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Mary A M Cleaton
- Department of Genetics, Downing Street, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Sarah Maurel
- IRSD, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Alice L Mitchell
- Department of Women and Children's Health, King's College London, Guy's Campus, London, UK
| | - Anne C Ferguson-Smith
- Department of Genetics, Downing Street, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Nicolas Cenac
- IRSD, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Catherine Williamson
- Department of Women and Children's Health, King's College London, Guy's Campus, London, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Marika Charalambous
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, SE19RT, UK.
- Pregnancy Physiology Laboratory, Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK.
| |
Collapse
|
11
|
Park JY, Kim HR, Lee SH, Lee SW, Sin HS, Kim SY, Park MH. Metabolic Profiling Changes Induced by Fermented Blackberries in High-Fat-Diet-Fed Mice Utilizing Gas Chromatography-Mass Spectrometry Analysis. BIOLOGY 2024; 13:511. [PMID: 39056704 PMCID: PMC11274121 DOI: 10.3390/biology13070511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/24/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024]
Abstract
The aim of this study was to investigate the metabolic changes associated with the anti-obesity effects of fermented blackberry extracts in the liver tissues of high-fat-diet-fed mice using mass spectrometry-based metabolomics analysis. C57BL/6J mice were divided into eight groups: normal-diet-fed mice, high-fat-diet-fed mice, high-fat diet treated with blackberry extract, high-fat-diet mice treated with blackberry fermented by L. plantarum, and high-fat diet with blackberry fermented by L. brevis. After 12 weeks, the high-fat-diet group exhibited a greater increase in liver weight compared to the control group, and among the groups, the group administered with blackberry fermented with L. plantarum showed the most pronounced reduction in liver weight. As the primary organ responsible for amino acid metabolism, the liver is crucial for maintaining amino acid homeostasis. In our study, we observed that the levels of several essential amino acids, including isoleucine and valine, were decreased by the high-fat diet, and were recovered by administration of blackberry extract fermented with L. plantarum. Our results demonstrated the potential of blackberry extract fermented with L. plantarum as a functional material for metabolic disorders by restoring some of the amino acid metabolism disturbances induced by a high-fat diet.
Collapse
Affiliation(s)
- Jae Young Park
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Deokjin-gu, Jeonju-si 54810, Jeonbuk State, Republic of Korea; (J.Y.P.); (H.-R.K.); (S.-H.L.)
| | - Ha-Rim Kim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Deokjin-gu, Jeonju-si 54810, Jeonbuk State, Republic of Korea; (J.Y.P.); (H.-R.K.); (S.-H.L.)
| | - Seung-Hyeon Lee
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Deokjin-gu, Jeonju-si 54810, Jeonbuk State, Republic of Korea; (J.Y.P.); (H.-R.K.); (S.-H.L.)
| | - Sang-Wang Lee
- Chebigen Inc., 62 Ballyong-ro, Deokjin-gu, Jeonju-si 54853, Jeonbuk State, Republic of Korea; (S.-W.L.); (H.-S.S.)
| | - Hong-Sig Sin
- Chebigen Inc., 62 Ballyong-ro, Deokjin-gu, Jeonju-si 54853, Jeonbuk State, Republic of Korea; (S.-W.L.); (H.-S.S.)
| | - Seon-Young Kim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Deokjin-gu, Jeonju-si 54810, Jeonbuk State, Republic of Korea; (J.Y.P.); (H.-R.K.); (S.-H.L.)
| | - Mi Hee Park
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Deokjin-gu, Jeonju-si 54810, Jeonbuk State, Republic of Korea; (J.Y.P.); (H.-R.K.); (S.-H.L.)
| |
Collapse
|
12
|
Centner AM, Shiel EA, Farra W, Cannon EN, Landim-Vieira M, Salazar G, Chelko SP. High-Fat Diet Augments Myocardial Inflammation and Cardiac Dysfunction in Arrhythmogenic Cardiomyopathy. Nutrients 2024; 16:2087. [PMID: 38999835 PMCID: PMC11243382 DOI: 10.3390/nu16132087] [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: 06/10/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a familial heart disease characterized by cardiac dysfunction, arrhythmias, and myocardial inflammation. Exercise and stress can influence the disease's progression. Thus, an investigation of whether a high-fat diet (HFD) contributes to ACM pathogenesis is warranted. In a robust ACM mouse model, 8-week-old Desmoglein-2 mutant (Dsg2mut/mut) mice were fed either an HFD or rodent chow for 8 weeks. Chow-fed wildtype (WT) mice served as controls. Echo- and electrocardiography images pre- and post-dietary intervention were obtained, and the lipid burden, inflammatory markers, and myocardial fibrosis were assessed at the study endpoint. HFD-fed Dsg2mut/mut mice showed numerous P-wave perturbations, reduced R-amplitude, left ventricle (LV) remodeling, and reduced ejection fraction (%LVEF). Notable elevations in plasma high-density lipoprotein (HDL) were observed, which correlated with the %LVEF. The myocardial inflammatory adipokines, adiponectin (AdipoQ) and fibroblast growth factor-1, were substantially elevated in HFD-fed Dsg2mut/mut mice, albeit no compounding effect was observed in cardiac fibrosis. The HFD not only potentiated cardiac dysfunction but additionally promoted adverse cardiac remodeling. Further investigation is warranted, particularly given elevated AdipoQ levels and the positive correlation of HDL with the %LVEF, which may suggest a protective effect. Altogether, the HFD worsened some, but not all, disease phenotypes in Dsg2mut/mut mice. Notwithstanding, diet may be a modifiable environmental factor in ACM disease progression.
Collapse
Affiliation(s)
- Ann M Centner
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Emily A Shiel
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Waleed Farra
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Elisa N Cannon
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Gloria Salazar
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Center for Advancing Exercise and Nutrition Research on Aging (CAENRA), Tallahassee, FL 32306, USA
| | - Stephen P Chelko
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| |
Collapse
|
13
|
Muzammil K, Hooshiar MH, Varmazyar S, Omar TM, Karim MM, Aadi S, Kalavi S, Yasamineh S. Potential use of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition and prevention method in viral infection. Microb Cell Fact 2024; 23:90. [PMID: 38528584 PMCID: PMC10962113 DOI: 10.1186/s12934-024-02355-8] [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: 01/09/2024] [Accepted: 03/03/2024] [Indexed: 03/27/2024] Open
Abstract
Cellular lipid membranes serve as the primary barrier preventing viral infection of the host cell and provide viruses with a critical initial point of contact. Occasionally, viruses can utilize lipids as viral receptors. Viruses depend significantly on lipid rafts for infection at virtually every stage of their life cycle. The pivotal role that proprotein convertase subtilisin/kexin Type 9 (PCSK9) plays in cholesterol homeostasis and atherosclerosis, primarily by post-transcriptionally regulating hepatic low-density lipoprotein receptor (LDLR) and promoting its lysosomal degradation, has garnered increasing interest. Conversely, using therapeutic, fully humanized antibodies to block PCSK9 leads to a significant reduction in high LDL cholesterol (LDL-C) levels. The Food and Drug Administration (FDA) has approved PCSK9 inhibitors, including inclisiran (Leqvio®), alirocumab (Praluent), and evolocumab (Repatha). At present, active immunization strategies targeting PCSK9 present a compelling substitute for passive immunization through the administration of antibodies. In addition to the current inquiry into the potential therapeutic application of PCSK9 inhibition in human immunodeficiency virus (HIV)-infected patients for hyperlipidemia associated with HIV and antiretroviral therapy (ART), preclinical research suggests that PCSK9 may also play a role in inhibiting hepatitis C virus (HCV) replication. Furthermore, PCSK9 inhibition has been suggested to protect against dengue virus (DENV) potentially and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses. Recent evidence regarding the impact of PCSK9 on a variety of viral infections, including HCV, HIV, DENV, and SARS-CoV-2, is examined in this article. As a result, PCSK9 inhibitors and vaccines may serve as viable host therapies for viral infections, as our research indicates that PCSK9 is significantly involved in the pathogenesis of viral infections.
Collapse
Affiliation(s)
- Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Khamis Mushait Campus, Abha, KSA, Saudi Arabia
| | | | - Shirin Varmazyar
- Department of Medicine, Shahroud Islamic azad university of medical sciences, Sharoud, Iran
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | - Manal Morad Karim
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Sadeq Aadi
- College of Dentistry, Al-Mustaqbal University, Babylon, 51001, Iraq
| | - Shaylan Kalavi
- Department of Clinical Pharmacy, faculty of pharmacy, Islamic Azad University of Medical Sciences, Tehran, Iran.
| | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| |
Collapse
|
14
|
Lund J, Lähteenmäki E, Eklund T, Bakke HG, Thoresen GH, Pirinen E, Jauhiainen M, Rustan AC, Lehti M. Human HDL subclasses modulate energy metabolism in skeletal muscle cells. J Lipid Res 2024; 65:100481. [PMID: 38008260 PMCID: PMC10770614 DOI: 10.1016/j.jlr.2023.100481] [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: 08/30/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023] Open
Abstract
In addition to its antiatherogenic role, HDL reportedly modulates energy metabolism at the whole-body level. HDL functionality is associated with its structure and composition, and functional activities can differ between HDL subclasses. Therefore, we studied if HDL2 and HDL3, the two major HDL subclasses, are able to modulate energy metabolism of skeletal muscle cells. Differentiated mouse and primary human skeletal muscle myotubes were used to investigate the influences of human HDL2 and HDL3 on glucose and fatty uptake and oxidation. HDL-induced changes in lipid distribution and mRNA expression of genes related to energy substrate metabolism, mitochondrial function, and HDL receptors were studied with human myotubes. Additionally, we examined the effects of apoA-I and discoidal, reconstituted HDL particles on substrate metabolism. In mouse myotubes, HDL subclasses strongly enhanced glycolysis upon high and low glucose concentrations. HDL3 caused a minor increase in ATP-linked respiration upon glucose conditioning but HDL2 improved complex I-mediated mitochondrial respiration upon fatty acid treatment. In human myotubes, glucose metabolism was attenuated but fatty acid uptake and oxidation were markedly increased by both HDL subclasses, which also increased mRNA expression of genes related to fatty acid metabolism and HDL receptors. Finally, both HDL subclasses induced incorporation of oleic acid into different lipid classes. These results, demonstrating that HDL subclasses enhance fatty acid oxidation in human myotubes but improve anaerobic metabolism in mouse myotubes, support the role of HDL as a circulating modulator of energy metabolism. Exact mechanisms and components of HDL causing the change, require further investigation.
Collapse
Affiliation(s)
- Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Emilia Lähteenmäki
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Tiia Eklund
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Hege G Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Research Unit for Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Matti Jauhiainen
- Department of Public Health and Welfare, Minerva Foundation Institute for Medical Research and Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Arild C Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Maarit Lehti
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
15
|
Sachan V, Le Dévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Resiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQH, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, Seidah NG. PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease. Metabolism 2024; 150:155736. [PMID: 37967646 DOI: 10.1016/j.metabol.2023.155736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Epidemiological evidence links the proprotein convertase subtilisin/kexin 7 (PCSK7) to triglyceride (TG) metabolism. We associated the known PCSK7 gain-of-function non-coding SNP rs236918 with higher levels of plasma apolipoprotein B (apoB) and the loss-of-function coding variant p.Pro777Leu (SNP rs201598301) with lower apoB and TG. Herein, we aimed to unravel the in vivo role of liver PCSK7. METHODS We biochemically defined the functional role of PCSK7 in lipid metabolism using hepatic cell lines and Pcsk7-/- mice. Our findings were validated following subcutaneous administration of hepatocyte-targeted N-acetylgalactosamine (GalNAc)-antisense oligonucleotides (ASOs) against Pcsk7. RESULTS Independent of its proteolytic activity, membrane-bound PCSK7 binds apoB100 in the endoplasmic reticulum and enhances its secretion. Mechanistically, the loss of PCSK7/Pcsk7 leads to apoB100 degradation, triggering an unfolded protein response, autophagy, and β-oxidation, eventually reducing lipid accumulation in hepatocytes. Non-alcoholic fatty liver disease (NAFLD) was induced by a 12-week high fat/fructose/cholesterol diet in wild type (WT) and Pcsk7-/- mice that were then allowed to recover on a 4-week control diet. Pcsk7-/- mice recovered more effectively than WT mice from all NAFLD-related liver phenotypes. Finally, subcutaneous administration of GalNAc-ASOs targeting hepatic Pcsk7 to WT mice validated the above results. CONCLUSIONS Our data reveal hepatic PCSK7 as one of the major regulators of apoB, and its absence reduces apoB secretion from hepatocytes favoring its ubiquitination and degradation by the proteasome. This results in a cascade of events, eventually reducing hepatic lipid accumulation, thus supporting the notion of silencing PCSK7 mRNA in hepatocytes for targeting NAFLD.
Collapse
Affiliation(s)
- Vatsal Sachan
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Maïlys Le Dévéhat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Anna Roubtsova
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Rachid Essalmani
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Jean-Francois Laurendeau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Damien Garçon
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Delia Susan-Resiga
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Stéphanie Duval
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Sahar Mikaeeli
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Josée Hamelin
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Alexandra Evagelidis
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Michael Chong
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | | | - Zu-Hua Gao
- Department of Pathology, McGill University Health Centre, Montréal, QC, Canada
| | - Isabelle Robillard
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Matthieu Ruiz
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Vincent Quoc-Huy Trinh
- Departement of Pathology and Cellular Biology, Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, QC, Canada
| | - Jennifer L Estall
- Molecular Mechanisms of Diabetes, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - May Faraj
- Nutrition Department, Université de Montréal, Research Unit on Nutrition, Lipoproteins and Cardiometabolic Diseases, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, Canada
| | - Martin Sauvageau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Annik Prat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Robert S Kiss
- McGill University Health Centre Research Institute, Montréal, QC, Canada
| | - Nabil G Seidah
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
16
|
Hernandez-Baixauli J, Chomiciute G, Alcaide-Hidalgo JM, Crescenti A, Baselga-Escudero L, Palacios-Jordan H, Foguet-Romero E, Pedret A, Valls RM, Solà R, Mulero M, Del Bas JM. Developing a model to predict the early risk of hypertriglyceridemia based on inhibiting lipoprotein lipase (LPL): a translational study. Sci Rep 2023; 13:22646. [PMID: 38114521 PMCID: PMC10730820 DOI: 10.1038/s41598-023-49277-w] [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: 09/20/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023] Open
Abstract
Hypertriglyceridemia (HTG) is an independent risk factor for atherosclerotic cardiovascular disease (ASCVD). One of the multiple origins of HTG alteration is impaired lipoprotein lipase (LPL) activity, which is an emerging target for HTG treatment. We hypothesised that early, even mild, alterations in LPL activity might result in an identifiable metabolomic signature. The aim of the present study was to assess whether a metabolic signature of altered LPL activity in a preclinical model can be identified in humans. A preclinical LPL-dependent model of HTG was developed using a single intraperitoneal injection of poloxamer 407 (P407) in male Wistar rats. A rat metabolomics signature was identified, which led to a predictive model developed using machine learning techniques. The predictive model was applied to 140 humans classified according to clinical guidelines as (1) normal, less than 1.7 mmol/L; (2) risk of HTG, above 1.7 mmol/L. Injection of P407 in rats induced HTG by effectively inhibiting plasma LPL activity. Significantly responsive metabolites (i.e. specific triacylglycerols, diacylglycerols, phosphatidylcholines, cholesterol esters and lysophospholipids) were used to generate a predictive model. Healthy human volunteers with the impaired predictive LPL signature had statistically higher levels of TG, TC, LDL and APOB than those without the impaired LPL signature. The application of predictive metabolomic models based on mechanistic preclinical research may be considered as a strategy to stratify subjects with HTG of different origins. This approach may be of interest for precision medicine and nutritional approaches.
Collapse
Affiliation(s)
- Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204, Reus, Spain
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gertruda Chomiciute
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204, Reus, Spain
| | | | - Anna Crescenti
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204, Reus, Spain
| | | | - Hector Palacios-Jordan
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204, Reus, Spain
| | - Elisabet Foguet-Romero
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204, Reus, Spain
| | - Anna Pedret
- Functional Nutrition, Oxidation and Cardiovascular Diseases Group (NFOC-Salut), Facultat de Medicina i Ciències de la Salut, Universitat Rovira I Virgili, C/Sant Llorenç, 21, 43201, Reus, Spain
| | - Rosa M Valls
- Functional Nutrition, Oxidation and Cardiovascular Diseases Group (NFOC-Salut), Facultat de Medicina i Ciències de la Salut, Universitat Rovira I Virgili, C/Sant Llorenç, 21, 43201, Reus, Spain
| | - Rosa Solà
- Functional Nutrition, Oxidation and Cardiovascular Diseases Group (NFOC-Salut), Facultat de Medicina i Ciències de la Salut, Universitat Rovira I Virgili, C/Sant Llorenç, 21, 43201, Reus, Spain
- Internal Medicine Service, Hospital Universitari Sant Joan de Reus, Av/del Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007, Tarragona, Spain.
| | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Àrea Biotecnologia, Reus, Spain.
| |
Collapse
|
17
|
Mitok KA, Schueler KL, King SM, Orr J, Ryan KA, Keller MP, Krauss RM, Mitchell BD, Shuldiner AR, Attie AD. Missense variants in SORT1 are associated with LDL-C in an Amish population. J Lipid Res 2023; 64:100468. [PMID: 37913995 PMCID: PMC10711479 DOI: 10.1016/j.jlr.2023.100468] [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: 08/09/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Common noncoding variants at the human 1p13.3 locus associated with SORT1 expression are among those most strongly associated with low-density lipoprotein cholesterol (LDL-C) in human genome-wide association studies. However, validation studies in mice and cell lines have produced variable results regarding the directionality of the effect of SORT1 on LDL-C. This, together with the fact that the 1p13.3 variants are associated with expression of several genes, has raised the question of whether SORT1 is the causal gene at this locus. Using whole exome sequencing in members of an Amish population, we identified coding variants in SORT1 that are associated with increased (rs141749679, K302E) and decreased (rs149456022, Q225H) LDL-C. Further, analysis of plasma lipoprotein particle subclasses by ion mobility in a subset of rs141749679 (K302E) carriers revealed higher levels of large LDL particles compared to noncarriers. In contrast to the effect of these variants in the Amish, the sortilin K302E mutation introduced into a C57BL/6J mouse via CRISPR/Cas9 resulted in decreased non-high-density lipoprotein cholesterol, and the sortilin Q225H mutation did not alter cholesterol levels in mice. This is indicative of different effects of these mutations on cholesterol metabolism in the two species. To our knowledge, this is the first evidence that naturally occurring coding variants in SORT1 are associated with LDL-C, thus supporting SORT1 as the gene responsible for the association of the 1p13.3 locus with LDL-C.
Collapse
Affiliation(s)
- Kelly A Mitok
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Kathryn L Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Sarah M King
- Department of Pediatrics, University of California-San Francisco, San Francisco, CA, USA
| | - Joseph Orr
- Department of Pediatrics, University of California-San Francisco, San Francisco, CA, USA
| | - Kathleen A Ryan
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ronald M Krauss
- Department of Pediatrics, University of California-San Francisco, San Francisco, CA, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan R Shuldiner
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Regeneron Genetics Center, Tarrytown, NY, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
18
|
Kessel D. Critical PDT Theory VII: Preclinical Translation. Photochem Photobiol 2023; 99:1361-1363. [PMID: 36862007 DOI: 10.1111/php.13797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/03/2023]
Abstract
The translation of photodynamic effects into clinical practice is a complex process that involves the pharmacokinetics of photosensitizing agents, light dosimetry and oxygenation levels. But even the 'translation' of basic photobiology into meaningful preclinical information can be challenging. Some thoughts on directions for progress in clinical trials are suggested.
Collapse
Affiliation(s)
- David Kessel
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
19
|
van Zwol W, Rimbert A, Wolters JC, Smit M, Bloks VW, Kloosterhuis NJ, Huijkman NCA, Koster MH, Tharehalli U, de Neck SM, Bournez C, Fuh MM, Kuipers J, Rajan S, de Bruin A, Ginsberg HN, van Westen GJP, Hussain MM, Scheja L, Heeren J, Zimmerman P, van de Sluis B, Kuivenhoven JA. Loss of hepatic SMLR1 causes hepatosteatosis and protects against atherosclerosis due to decreased hepatic VLDL secretion. Hepatology 2023; 78:1418-1432. [PMID: 36053190 PMCID: PMC10581432 DOI: 10.1002/hep.32709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS The assembly and secretion of VLDL from the liver, a pathway that affects hepatic and plasma lipids, remains incompletely understood. We set out to identify players in the VLDL biogenesis pathway by identifying genes that are co-expressed with the MTTP gene that encodes for microsomal triglyceride transfer protein, key to the lipidation of apolipoprotein B, the core protein of VLDL. Using human and murine transcriptomic data sets, we identified small leucine-rich protein 1 ( SMLR1 ), encoding for small leucine-rich protein 1, a protein of unknown function that is exclusively expressed in liver and small intestine. APPROACH AND RESULTS To assess the role of SMLR1 in the liver, we used somatic CRISPR/CRISPR-associated protein 9 gene editing to silence murine Smlr1 in hepatocytes ( Smlr1 -LKO). When fed a chow diet, male and female mice show hepatic steatosis, reduced plasma apolipoprotein B and triglycerides, and reduced VLDL secretion without affecting microsomal triglyceride transfer protein activity. Immunofluorescence studies show that SMLR1 is in the endoplasmic reticulum and Cis-Golgi complex. The loss of hepatic SMLR1 in female mice protects against diet-induced hyperlipidemia and atherosclerosis but causes NASH. On a high-fat, high-cholesterol diet, insulin and glucose tolerance tests did not reveal differences in male Smlr1 -LKO mice versus controls. CONCLUSIONS We propose a role for SMLR1 in the trafficking of VLDL from the endoplasmic reticulum to the Cis-Golgi complex. While this study uncovers SMLR1 as a player in the VLDL assembly, trafficking, and secretion pathway, it also shows that NASH can occur with undisturbed glucose homeostasis and atheroprotection.
Collapse
Affiliation(s)
- Willemien van Zwol
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Antoine Rimbert
- Université de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Justina C. Wolters
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marieke Smit
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Vincent W. Bloks
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nicolette C. A. Huijkman
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mirjam H. Koster
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Umesh Tharehalli
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Simon M. de Neck
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Colin Bournez
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Marceline M. Fuh
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg−Eppendorf, Hamburg, Germany
| | - Jeroen Kuipers
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sujith Rajan
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Alain de Bruin
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Gerard J. P. van Westen
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg−Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg−Eppendorf, Hamburg, Germany
| | | | - Bart van de Sluis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
20
|
Madigan V, Zhang F, Dahlman JE. Drug delivery systems for CRISPR-based genome editors. Nat Rev Drug Discov 2023; 22:875-894. [PMID: 37723222 DOI: 10.1038/s41573-023-00762-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 09/20/2023]
Abstract
CRISPR-based drugs can theoretically manipulate any genetic target. In practice, however, these drugs must enter the desired cell without eliciting an unwanted immune response, so a delivery system is often required. Here, we review drug delivery systems for CRISPR-based genome editors, focusing on adeno-associated viruses and lipid nanoparticles. After describing how these systems are engineered and their subsequent characterization in preclinical animal models, we highlight data from recent clinical trials. Preclinical targeting mediated by polymers, proteins, including virus-like particles, and other vehicles that may deliver CRISPR systems in the future is also discussed.
Collapse
Affiliation(s)
- Victoria Madigan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Feng Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research at MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
21
|
Brenner EP, Sreevatsan S. Global-scale GWAS associates a subset of SNPs with animal-adapted variants in M. tuberculosis complex. BMC Med Genomics 2023; 16:260. [PMID: 37875894 PMCID: PMC10598944 DOI: 10.1186/s12920-023-01695-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND While Mycobacterium tuberculosis complex (MTBC) variants are clonal, variant tuberculosis is a human-adapted pathogen, and variant bovis infects many hosts. Despite nucleotide identity between MTBC variants exceeding 99.95%, it remains unclear what drives these differences. Markers of adaptation into variants were sought by bacterial genome-wide association study of single nucleotide polymorphisms extracted from 6,362 MTBC members from varied hosts and countries. RESULTS The search identified 120 genetic loci associated with MTBC variant classification and certain hosts. In many cases, these changes are uniformly fixed in certain variants while absent in others in this dataset, providing good discriminatory power in distinguishing variants by polymorphisms. Multiple changes were seen in genes for cholesterol and fatty acid metabolism, pathways previously proposed to be important for host adaptation, including Mce4F (part of the fundamental cholesterol intake Mce4 pathway), 4 FadD and FadE genes (playing roles in cholesterol and fatty acid utilization), and other targets like Rv3548c and PTPB, genes shown essential for growth on cholesterol by transposon studies. CONCLUSIONS These findings provide a robust set of genetic loci associated with the split of variant bovis and variant tuberculosis, and suggest that adaptation to new hosts could involve adjustments in uptake and catabolism of cholesterol and fatty acids, like the proposed specialization to different populations in MTB lineages by alterations to host lipid composition. Future studies are required to elucidate how the associations between cholesterol profiles and pathogen utilization differences between hosts and MTBC variants, as well as the investigation of uncharacterized genes discovered in this study. This information will likely provide an understanding on the diversification of MBO away from humans and specialization towards a broad host range.
Collapse
Affiliation(s)
- Evan P Brenner
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, 784 Wilson Road, East Lansing, MI, 48824, USA
| | - Srinand Sreevatsan
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, 784 Wilson Road, East Lansing, MI, 48824, USA.
| |
Collapse
|
22
|
Dai W, Castleberry M, Zheng Z. Tale of two systems: the intertwining duality of fibrinolysis and lipoprotein metabolism. J Thromb Haemost 2023; 21:2679-2696. [PMID: 37579878 PMCID: PMC10599797 DOI: 10.1016/j.jtha.2023.08.004] [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: 03/16/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023]
Abstract
Fibrinolysis is an enzymatic process that breaks down fibrin clots, while dyslipidemia refers to abnormal levels of lipids and lipoproteins in the blood. Both fibrinolysis and lipoprotein metabolism are critical mechanisms that regulate a myriad of functions in the body, and the imbalance of these mechanisms is linked to the development of pathologic conditions, such as thrombotic complications in atherosclerotic cardiovascular diseases. Accumulated evidence indicates the close relationship between the 2 seemingly distinct and complicated systems-fibrinolysis and lipoprotein metabolism. Observational studies in humans found that dyslipidemia, characterized by increased blood apoB-lipoprotein and decreased high-density lipoprotein, is associated with lower fibrinolytic potential. Genetic variants of some fibrinolytic regulators are associated with blood lipid levels, supporting a causal relationship between these regulators and lipoprotein metabolism. Mechanistic studies have elucidated many pathways that link the fibrinolytic system and lipoprotein metabolism. Moreover, profibrinolytic therapies improve lipid panels toward an overall cardiometabolic healthier phenotype, while some lipid-lowering treatments increase fibrinolytic potential. The complex relationship between lipoprotein and fibrinolysis warrants further research to improve our understanding of the bidirectional regulation between the mediators of fibrinolysis and lipoprotein metabolism.
Collapse
Affiliation(s)
- Wen Dai
- Versiti Blood Research Institute, Milwaukee, USA.
| | | | - Ze Zheng
- Versiti Blood Research Institute, Milwaukee, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, USA.
| |
Collapse
|
23
|
van Dijk RA, Kleemann R, Schaapherder AF, van den Bogaerdt A, Hedin U, Matic L, Lindeman JH. Validating human and mouse tissues commonly used in atherosclerosis research with coronary and aortic reference tissue: similarities but profound differences in disease initiation and plaque stability. JVS Vasc Sci 2023; 4:100118. [PMID: 37810738 PMCID: PMC10551657 DOI: 10.1016/j.jvssci.2023.100118] [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: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 10/10/2023] Open
Abstract
Objective Characterization of the atherosclerotic process fully relies on histological evaluation and staging through a consensus grading system. So far, a head-to-head comparison of atherosclerotic process in experimental models and tissue resources commonly applied in atherosclerosis research with the actual human atherosclerotic process is missing. Material and Methods Aspects of the atherosclerotic process present in established murine atherosclerosis models and human carotid endarterectomy specimen were systematically graded using the modified American Heart Association histological classification (Virmani classification). Aspects were aligned with the atherosclerotic process observed in human coronary artery and aortic atherosclerosis reference tissues that were available through biobanks based on human tissue/organ donor material. Results Apart from absent intraplaque hemorrhages in aortic lesions, the histological characteristics of the different stages of human coronary and aortic atherosclerosis are similar. Carotid endarterectomy samples all represent end-stage "fibrous calcified plaque" lesions, although secondary, progressive, and vulnerable lesions with gross morphologies similar to coronary/aortic lesions occasionally present along the primary lesions. For the murine lesions, clear histological parallels were observed for the intermediate lesion types ("pathological intimal thickening," and "early fibroatheroma"). However, none of the murine lesions studied progressed to an equivalent of late fibroatheroma or beyond. Notable contrasts were observed for disease initiation: whereas disease initiation in humans is characterized by a mesenchymal cell influx in the intima, the earliest murine lesions are exclusively intimal, with subendothelial accumulation foam cells. A mesenchymal (and medial) response are absent. In fact, it is concluded that the stage of "adaptive intimal thickening" is absent in all mouse models included in this study. Conclusions The Virmani classification for coronary atherosclerosis can be applied for systematically grading experimental and clinical atherosclerosis. Application of this histological grading tool shows clear parallels for intermediate human and murine atherosclerotic lesions. However, clear contrasts are observed for disease initiation, and late stage atherosclerotic lesions. Carotid endarterectomy all represent end-stage fibrous calcified plaque lesions, although secondary earlier lesions may present in a subset of samples.
Collapse
Affiliation(s)
- Rogier A. van Dijk
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Kleemann
- The Netherlands Organization for Applied Scientific Research (TNO), Department of Metabolic Health Research, TNO Metabolic Health Research, Leiden, The Netherlands
| | | | | | - Ulf Hedin
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Ljubica Matic
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Jan H.N. Lindeman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Abstract
All mammalian cell membranes contain cholesterol to maintain membrane integrity. The transport of this hydrophobic lipid is mediated by lipoproteins. Cholesterol is especially enriched in the brain, particularly in synaptic and myelin membranes. Aging involves changes in sterol metabolism in peripheral organs and also in the brain. Some of those alterations have the potential to promote or to counteract the development of neurodegenerative diseases during aging. Here, we summarize the current knowledge of general principles of sterol metabolism in humans and mice, the most widely used model organism in biomedical research. We discuss changes in sterol metabolism that occur in the aged brain and highlight recent developments in cell type-specific cholesterol metabolism in the fast-growing research field of aging and age-related diseases, focusing on Alzheimer's disease. We propose that cell type-specific cholesterol handling and the interplay between cell types critically influence age-related disease processes.
Collapse
Affiliation(s)
- Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany;
| |
Collapse
|
25
|
Gautam J, Kumari D, Aggarwal H, Gupta SK, Kasarla SS, Sarkar S, Priya MRK, Kamboj P, Kumar Y, Dikshit M. Characterization of lipid signatures in the plasma and insulin-sensitive tissues of the C57BL/6J mice fed on obesogenic diets. Biochim Biophys Acta Mol Cell Biol Lipids 2023:159348. [PMID: 37285928 DOI: 10.1016/j.bbalip.2023.159348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Diet-induced obesity mouse models are widely utilized to investigate the underlying mechanisms of dyslipidemia, glucose intolerance, insulin resistance, hepatic steatosis, and type 2 diabetes mellitus (T2DM), as well as for screening potential drug compounds. However, there is limited knowledge regarding specific signature lipids that accurately reflect dietary disorders. In this study, we aimed to identify key lipid signatures using LC/MS-based untargeted lipidomics in the plasma, liver, adipose tissue (AT), and skeletal muscle tissues (SKM) of male C57BL/6J mice that were fed chow, LFD, or obesogenic diets (HFD, HFHF, and HFCD) for a duration of 20 weeks. Furthermore, we conducted a comprehensive lipid analysis to assess similarities and differences with human lipid profiles. The mice fed obesogenic diets exhibited weight gain, glucose intolerance, elevated BMI, glucose and insulin levels, and a fatty liver, resembling characteristics of T2DM and obesity in humans. In total, we identified approximately 368 lipids in plasma, 433 in the liver, 493 in AT, and 624 in SKM. Glycerolipids displayed distinct patterns across the tissues, differing from human findings. However, changes in sphingolipids, phospholipids, and the expression of inflammatory and fibrotic genes showed similarities to reported human findings. Significantly modulated pathways in the obesogenic diet-fed groups included ceramide de novo synthesis, sphingolipid remodeling, and the carboxylesterase pathway, while lipoprotein-mediated pathways were minimally affected.
Collapse
Affiliation(s)
- Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Hobby Aggarwal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Siva Swapna Kasarla
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Soumalya Sarkar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - M R Kamla Priya
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Parul Kamboj
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| | - Madhu Dikshit
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
26
|
Khalikova D, An'kov S, Zhukova N, Tolstikova T, Popov S, Saiko A. Effect of the Composition of Leuzea and Cranberry Meal Extracts on Metabolic Processes in Norm and Pathology. Pharmaceuticals (Basel) 2023; 16:ph16050768. [PMID: 37242551 DOI: 10.3390/ph16050768] [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: 04/17/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
This study was conducted to evaluate the effects of long-term administration of a new herbal composition of leuzea and cranberry meal extracts at a dose of 70:500 mg/kg in healthy and pathological mice. After 4 weeks of daily composition administration to healthy CD-1 mice and C57BL/6 mice with diet-induced metabolic syndrome, oral glucose tolerance test (OGTT), serum biochemical examination and histology of internal organs were performed. Additionally, histological examination of white and brown adipose tissue was performed to evaluate the ability of the composition to prevent abdominal obesity in C57BL/6Ay (agouti yellow) mice. The results showed that the composition increased tissue sensitivity to glucose in healthy CD-1 mice; at the same time, it did not worsen the course of pathological processes in pathological mice. In both cases, the application of the developed composition was safe and contributed to the restoration of metabolic parameters.
Collapse
Affiliation(s)
- Daria Khalikova
- Vorozhtzov Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Science, 9 Akademika Lavrentieva Ave., Novosibirsk 630090, Russia
| | - Sergey An'kov
- Vorozhtzov Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Science, 9 Akademika Lavrentieva Ave., Novosibirsk 630090, Russia
| | - Nataliya Zhukova
- Vorozhtzov Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Science, 9 Akademika Lavrentieva Ave., Novosibirsk 630090, Russia
| | - Tatyana Tolstikova
- Vorozhtzov Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Science, 9 Akademika Lavrentieva Ave., Novosibirsk 630090, Russia
| | - Sergey Popov
- Vorozhtzov Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Science, 9 Akademika Lavrentieva Ave., Novosibirsk 630090, Russia
| | - Anastasia Saiko
- Vorozhtzov Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Science, 9 Akademika Lavrentieva Ave., Novosibirsk 630090, Russia
| |
Collapse
|
27
|
Woo JG, Melchior JT, Swertfeger DK, Remaley AT, Sise EA, Sosseh F, Welge JA, Prentice AM, Davidson WS, Moore SE, Woollett LA. Lipoprotein subfraction patterns throughout gestation in The Gambia: changes in subfraction composition and their relationships with infant birth weights. Lipids Health Dis 2023; 22:19. [PMID: 36737730 PMCID: PMC9896684 DOI: 10.1186/s12944-023-01776-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Lipoprotein subfraction concentrations have been shown to change as gestation progresses in resource-rich settings. The objective of the current study was to evaluate the impact of pregnancy on different-sized lipoprotein particle concentrations and compositions in a resource-poor setting. METHOD Samples were collected from pregnant women in rural Gambia at enrollment (8-20 weeks), 20 weeks, and 30 weeks of gestation. Concentrations of different-sized high-density, low-density, and triglyceride-rich lipoprotein particles (HDL, LDL, and TRL, respectively) were measured by nuclear magnetic resonance in 126 pooled plasma samples from a subset of women. HDL was isolated and the HDL proteome evaluated using mass spectroscopy. Subfraction concentrations from women in The Gambia were also compared to concentrations in women in the U.S. in mid gestation. RESULTS Total lipoprotein particles and all-sized TRL, LDL, and HDL particle concentrations increased during gestation, with the exception of medium-sized LDL and HDL particles which decreased. Subfraction concentrations were not associated with infant birth weights, though relationships were found between some lipoprotein subfraction concentrations in women with normal versus low birth weight infants (< 2500 kg). HDL's proteome also changed during gestation, showing enrichment in proteins associated with metal ion binding, hemostasis, lipid metabolism, protease inhibitors, proteolysis, and complement activation. Compared to women in the U.S., Gambian women had lower large- and small-sized LDL and HDL concentrations, but similar medium-sized LDL and HDL concentrations. CONCLUSIONS Most lipoprotein subfraction concentrations increase throughout pregnancy in Gambian women and are lower in Gambian vs U.S. women, the exception being medium-sized LDL and HDL particle concentrations which decrease during gestation and are similar in both cohorts of women. The proteomes of HDL also change in ways to support gestation. These changes warrant further study to determine how a lack of change or different changes could impact negative pregnancy outcomes.
Collapse
Affiliation(s)
- Jessica G Woo
- Departments of Pediatrics and Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - John T Melchior
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Biological Sciences Division, Pacific Northwest National Laboratory, WA, Richland, USA
| | - Debi K Swertfeger
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ebrima A Sise
- MRC Unit The Gambia, London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Fatou Sosseh
- MRC Unit The Gambia, London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Jeffrey A Welge
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Andrew M Prentice
- MRC Unit The Gambia, London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sophie E Moore
- MRC Unit The Gambia, London School of Hygiene and Tropical Medicine, Banjul, The Gambia
- Department of Women and Children's Health, King's College London, London, UK
| | - Laura A Woollett
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
28
|
Yuzbashian E, Moftah S, Chan CB. Graduate Student Literature Review: A scoping review on the impact of consumption of dairy products on phosphatidylcholine and lysophosphatidylcholine in circulation and the liver in human studies and animal models. J Dairy Sci 2023; 106:24-38. [PMID: 36400621 DOI: 10.3168/jds.2022-21938] [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: 02/07/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022]
Abstract
Dairy consumption is inversely related to the risk of developing type 2 diabetes in epidemiological research. One proposed hypothesis is that phospholipid (PL) species associated with dairy consumption mediate this relationship. This scoping review aimed to identify the existing literature in animal and human trials investigating the impact of dairy products, including milk, yogurt, and cheese as well as dairy-derived PL supplementation on PL and its species in the circulation, summarizing the characteristics of these studies and identifying research gaps. A systematic search was conducted across 3 databases (PubMed, Scopus, and Web of Science) in March 2021. Of 2,427 identified references, 15 studies (7 humans and 8 animal studies) met the eligibility criteria and were included in the final narrative synthesis. The evidence base was heterogeneous, involving a variety of clinical and preclinical studies, metabolically healthy or obese/diabetic participants or animal models, and displayed mixed findings. Circulating postprandial concentrations of total PL were elevated acutely but unchanged after longer intervention with dairy products. The PL concentration remained stable even after a high dosage of milk supplemented with dairy-derived PL, which may be related to increased fecal excretion; however, certain phosphatidylcholine (PC) or lysophosphatidylcholine species were increased in circulation by interventions. These include several PC species with 32 to 38 total carbons in addition to the dairy biomarkers C15:0 and C17:0. The results of this scoping review demonstrate a small body of literature indicating that dairy products can influence blood concentrations of PC and lysophosphatidylcholine species in both rodents and humans without alteration of total PL and PC. There is a lack of well-designed trials in humans and animals that explore the potential differences between individual dairy foods on PL species. In addition, trials to understand the bioactive properties of PC and lysophosphatidylcholine species on cardiometabolic risk are needed.
Collapse
Affiliation(s)
- Emad Yuzbashian
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5
| | - Salma Moftah
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Catherine B Chan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2P5; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada T6G 2H7.
| |
Collapse
|
29
|
Finney AC, Das S, Kumar D, McKinney MP, Cai B, Yurdagul A, Rom O. The interplay between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease. Front Cardiovasc Med 2023; 10:1116861. [PMID: 37200978 PMCID: PMC10185914 DOI: 10.3389/fcvm.2023.1116861] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/23/2023] [Indexed: 05/20/2023] Open
Abstract
Therapeutic approaches that lower circulating low-density lipoprotein (LDL)-cholesterol significantly reduced the burden of cardiovascular disease over the last decades. However, the persistent rise in the obesity epidemic is beginning to reverse this decline. Alongside obesity, the incidence of nonalcoholic fatty liver disease (NAFLD) has substantially increased in the last three decades. Currently, approximately one third of world population is affected by NAFLD. Notably, the presence of NAFLD and particularly its more severe form, nonalcoholic steatohepatitis (NASH), serves as an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), thus, raising interest in the relationship between these two diseases. Importantly, ASCVD is the major cause of death in patients with NASH independent of traditional risk factors. Nevertheless, the pathophysiology linking NAFLD/NASH with ASCVD remains poorly understood. While dyslipidemia is a common risk factor underlying both diseases, therapies that lower circulating LDL-cholesterol are largely ineffective against NASH. While there are no approved pharmacological therapies for NASH, some of the most advanced drug candidates exacerbate atherogenic dyslipidemia, raising concerns regarding their adverse cardiovascular consequences. In this review, we address current gaps in our understanding of the mechanisms linking NAFLD/NASH and ASCVD, explore strategies to simultaneously model these diseases, evaluate emerging biomarkers that may be useful to diagnose the presence of both diseases, and discuss investigational approaches and ongoing clinical trials that potentially target both diseases.
Collapse
Affiliation(s)
- Alexandra C. Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - M. Peyton McKinney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| |
Collapse
|
30
|
Mangu SR, Patel K, Sukhdeo SV, Savitha MR, Sharan K. Maternal high-cholesterol diet negatively programs offspring bone development and downregulates hedgehog signaling in osteoblasts. J Biol Chem 2022; 298:102324. [PMID: 35931113 PMCID: PMC9440389 DOI: 10.1016/j.jbc.2022.102324] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cholesterol is one of the essential intrauterine factors required for fetal growth and development. Maternal high cholesterol levels are known to be detrimental for offspring health. However, its long-term effect on offspring skeletal development remains to be elucidated. We performed our studies in two strains of mice (C57BL6/J and Swiss Albino) and human subjects (65 mother-female newborn dyads) to understand the regulation of offspring skeletal growth by maternal high cholesterol. We found that mice offspring from high-cholesterol-fed dams had low birth weight, smaller body length, and delayed skeletal ossification at the E18.5 embryonic stage. Moreover, we observed that the offspring did not recover from the reduced skeletal mass and exhibited a low bone mass phenotype throughout their life. We attributed this effect to reduced osteoblast cell activity with a concomitant increase in the osteoclast cell population. Our investigation of the molecular mechanism revealed that offspring from high-cholesterol-fed dams had a decrease in the expression of ligands and proteins involved in hedgehog signaling. Further, our cross-sectional study of human subjects showed a significant inverse correlation between maternal blood cholesterol levels and cord blood bone formation markers. Moreover, the bone formation markers were significantly lower in the female newborns of hypercholesterolemic mothers compared with mothers with normal cholesterolemic levels. Together, our results suggest that maternal high cholesterol levels deleteriously program offspring bone mass and bone quality and downregulate the hedgehog signaling pathway in their osteoblasts.
Collapse
Affiliation(s)
- Svvs Ravi Mangu
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kalpana Patel
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shinde Vijay Sukhdeo
- Department of Meat and Marine Sciences, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - M R Savitha
- Department of Paediatrics, Mysore Medical College and Research Institute, Mysuru, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
31
|
Ngo W, Wu JLY, Lin ZP, Zhang Y, Bussin B, Granda Farias A, Syed AM, Chan K, Habsid A, Moffat J, Chan WCW. Identifying cell receptors for the nanoparticle protein corona using genome screens. Nat Chem Biol 2022; 18:1023-1031. [PMID: 35953550 DOI: 10.1038/s41589-022-01093-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 06/22/2022] [Indexed: 01/01/2023]
Abstract
Nanotechnology provides platforms to deliver medical agents to specific cells. However, the nanoparticle's surface becomes covered with serum proteins in the blood after administration despite engineering efforts to protect it with targeting or blocking molecules. Here, we developed a strategy to identify the main interactions between nanoparticle-adsorbed proteins and a cell by integrating mass spectrometry with pooled genome screens and Search Tool for the Retrieval of Interacting Genes analysis. We found that the low-density lipoprotein (LDL) receptor was responsible for approximately 75% of serum-coated gold nanoparticle uptake in U-87 MG cells. Apolipoprotein B and complement C8 proteins on the nanoparticle mediated uptake through the LDL receptor. In vivo, nanoparticle accumulation correlated with LDL receptor expression in the organs of mice. A detailed understanding of how adsorbed serum proteins bind to cell receptors will lay the groundwork for controlling the delivery of nanoparticles at the molecular level to diseased tissues for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Wayne Ngo
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Jamie L Y Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Zachary P Lin
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Yuwei Zhang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Bram Bussin
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Granda Farias
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Abdullah M Syed
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Katherine Chan
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Habsid
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Jason Moffat
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. .,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada. .,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada. .,Department of Materials Science and Engineering, University of Toronto, Toronto, Ontario, Canada. .,Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
32
|
Xiang P, Blanchard V, Francis GA. Smooth Muscle Cell—Macrophage Interactions Leading to Foam Cell Formation in Atherosclerosis: Location, Location, Location. Front Physiol 2022; 13:921597. [PMID: 35795646 PMCID: PMC9251363 DOI: 10.3389/fphys.2022.921597] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Cholesterol-overloaded cells or “foam cells” in the artery wall are the biochemical hallmark of atherosclerosis, and are responsible for much of the growth, inflammation and susceptibility to rupture of atherosclerotic lesions. While it has previously been thought that macrophages are the main contributor to the foam cell population, recent evidence indicates arterial smooth muscle cells (SMCs) are the source of the majority of foam cells in both human and murine atherosclerosis. This review outlines the timeline, site of appearance and proximity of SMCs and macrophages with lipids in human and mouse atherosclerosis, and likely interactions between SMCs and macrophages that promote foam cell formation and removal by both cell types. An understanding of these SMC-macrophage interactions in foam cell formation and regression is expected to provide new therapeutic targets to reduce the burden of atherosclerosis for the prevention of coronary heart disease, stroke and peripheral vascular disease.
Collapse
|
33
|
Kim KR, Kim J, Back JH, Lee JE, Ahn DR. Cholesterol-Mediated Seeding of Protein Corona on DNA Nanostructures for Targeted Delivery of Oligonucleotide Therapeutics to Treat Liver Fibrosis. ACS NANO 2022; 16:7331-7343. [PMID: 35500062 DOI: 10.1021/acsnano.1c08508] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The protein corona is a protein layer formed on the surface of nanoparticles administered in vivo and considerably affects the in vivo fate of nanoparticles. Although it is challenging to control protein adsorption on nanoparticles precisely, the protein corona may be harnessed to develop a targeted drug delivery system if the nanoparticles are decorated with a ligand with enhanced affinity to target tissue- and cell-homing proteins. Here, we prepared a DNA tetrahedron with trivalent cholesterol conjugation (Chol3-Td) that can induce enhanced interaction with lipoproteins in serum, which in situ generates the lipoprotein-associated protein corona on a DNA nanostructure favorable for cells abundantly expressing lipoprotein receptors in the liver, such as hepatocytes in healthy mice and myofibroblasts in fibrotic mice. Chol3-Td was further adopted for liver delivery of antisense oligonucleotide (ASO) targeting TGF-β1 mRNA to treat liver fibrosis in a mouse model. The potency of ASO@Chol3-Td was comparable to that of ASO conjugated with the clinically approved liver-targeting ligand, trivalent N-acetylgalactosamine (GalNAc3), demonstrating the potential of Chol3-Td as a targeted delivery system for oligonucleotide therapeutics. This study suggests that controlled seeding of the protein corona on nanomaterials can provide a way to steer nanoparticles into the target area.
Collapse
Affiliation(s)
- Kyoung-Ran Kim
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Junghyun Kim
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Ji Hyun Back
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Ji Eun Lee
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Dae-Ro Ahn
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
- Division of Biomedical Science and Technology, KIST School, University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| |
Collapse
|
34
|
Ellis JL, Fu X, Karl JP, Hernandez CJ, Mason JB, DeBose-Boyd RA, Booth SL. Multiple Dietary Vitamin K Forms Are Converted to Tissue Menaquinone-4 in Mice. J Nutr 2022; 152:981-993. [PMID: 34550377 PMCID: PMC8971004 DOI: 10.1093/jn/nxab332] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/01/2021] [Accepted: 09/14/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Vitamin K is a term that comprises a family of structurally related quinones, phylloquinone (PK) and the menaquinones (MKn), that share a common naphthoquinone ring but vary in sidechain length (n) and saturation. Dietary PK is a biosynthetic precursor to tissue menaquinone-4 (MK4), but little is known about the absorption and metabolism of dietary MKn. OBJECTIVE To characterize the absorption and metabolism of dietary MKn relative to PK. METHODS In the 4-week diet study, 10-week-old male and female C57BL/6 mice were pair-fed a vitamin K deficient diet (control) or a diet supplemented with 5.0 μmol/kg total PK, MK4, and/or MK9 (separately and in combination). In the 1-week stable isotope study, 12-week-old mice were pair-fed diets containing 2.2 μmol/kg PK (unlabeled control), 2H7PK, 13C11MK4, 2H7MK7, or 2H7MK9. Vitamin K tissue content was quantified by HPLC and/or LC-MS, and concentrations were compared by sex and diet group using 2-factor ANOVA. RESULTS Regardless of the form(s) of vitamin K provided in the diet, tissue MK4 concentrations did not differ across equimolar supplemented groups in the kidney, adipose, reproductive organ, bone, or pancreas in either males or females in the diet study (all P values > 0.05). Isotopic labeling confirmed the naphthoquinone ring of MK4 in tissues originated from the administered dietary PK or MKn. Despite equimolar supplementation, accumulation of the administered dietary form differed across diet groups in small intestinal segments (all P values < 0.002) and the liver (P < 0.001). Female mice had greater total vitamin K than males in every tissue examined (P < 0.05). CONCLUSIONS Dietary PK, MK4, MK7, and MK9 all served as precursors to tissue MK4 in mice. This study expands our understanding of vitamin K metabolism and supports a common conversion mechanism of all dietary vitamin K forms to MK4. Further investigation of the metabolism and physiological roles of MK4 that may be independent of classical vitamin K function is warranted.
Collapse
Affiliation(s)
- Jessie L Ellis
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
- The Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA
| | - Xueyan Fu
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - J Philip Karl
- US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Christopher J Hernandez
- Schools of Mechanical and Aerospace Engineering & Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Joel B Mason
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Russell A DeBose-Boyd
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarah L Booth
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| |
Collapse
|
35
|
Luo Y, Lu H, Peng D, Ruan X, Chen YE, Guo Y. Liver-humanized mice: A translational strategy to study metabolic disorders. J Cell Physiol 2022; 237:489-506. [PMID: 34661916 PMCID: PMC9126562 DOI: 10.1002/jcp.30610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 01/03/2023]
Abstract
The liver is the metabolic core of the whole body. Tools commonly used to study the human liver metabolism include hepatocyte cell lines, primary human hepatocytes, and pluripotent stem cells-derived hepatocytes in vitro, and liver genetically humanized mouse model in vivo. However, none of these systems can mimic the human liver in physiological and pathological states satisfactorily. Liver-humanized mice, which are established by reconstituting mouse liver with human hepatocytes, have emerged as an attractive animal model to study drug metabolism and evaluate the therapeutic effect in "human liver" in vivo because the humanized livers greatly replicate enzymatic features of human hepatocytes. The application of liver-humanized mice in studying metabolic disorders is relatively less common due to the largely uncertain replication of metabolic profiles compared to humans. Here, we summarize the metabolic characteristics and current application of liver-humanized mouse models in metabolic disorders that have been reported in the literature, trying to evaluate the pros and cons of using liver-humanized mice as novel mouse models to study metabolic disorders.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangbo Ruan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins School of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Center for Advanced Models and Translational Sciences and Therapeutics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
36
|
Landowski M, Bowes Rickman C. Targeting Lipid Metabolism for the Treatment of Age-Related Macular Degeneration: Insights from Preclinical Mouse Models. J Ocul Pharmacol Ther 2021; 38:3-32. [PMID: 34788573 PMCID: PMC8817708 DOI: 10.1089/jop.2021.0067] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) is a major leading cause of irreversible visual impairment in the world with limited therapeutic interventions. Histological, biochemical, genetic, and epidemiological studies strongly implicate dysregulated lipid metabolism in the retinal pigmented epithelium (RPE) in AMD pathobiology. However, effective therapies targeting lipid metabolism still need to be identified and developed for this blinding disease. To test lipid metabolism-targeting therapies, preclinical AMD mouse models are needed to establish therapeutic efficacy and the role of lipid metabolism in the development of AMD-like pathology. In this review, we provide a comprehensive overview of current AMD mouse models available to researchers that could be used to provide preclinical evidence supporting therapies targeting lipid metabolism for AMD. Based on previous studies of AMD mouse models, we discuss strategies to modulate lipid metabolism as well as examples of studies evaluating lipid-targeting therapeutics to restore lipid processing in the RPE. The use of AMD mouse models may lead to worthy lipid-targeting candidate therapies for clinical trials to prevent the blindness caused by AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
37
|
Kaesler N, Schreibing F, Speer T, Puente-Secades SDL, Rapp N, Drechsler C, Kabgani N, Kuppe C, Boor P, Jankowski V, Schurgers L, Kramann R, Floege J. Altered vitamin K biodistribution and metabolism in experimental and human chronic kidney disease. Kidney Int 2021; 101:338-348. [PMID: 34774554 DOI: 10.1016/j.kint.2021.10.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/07/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022]
Abstract
Chronic kidney disease (CKD) is accompanied with extensive cardiovascular calcification, in part correlating with functional vitamin K deficiency. Here, we sought to determine causes for vitamin K deficiency beyond reduced dietary intake. Initially, vitamin K uptake and distribution into circulating lipoproteins after a single administration of vitamin K1 plus K2 (menaquinone 4 and menaquinone 7, respectively) was determined in patients on dialysis therapy and healthy individuals. The patients incorporated very little menaquinone 7 but more menaquinone 4 into high density lipoprotein (HDL) and low-density lipoprotein particles than did healthy individuals. In contrast to healthy persons, HDL particles from the patients could not be spiked with menaquinone 7 in vitro and HDL uptake was diminished in osteoblasts. A reduced carboxylation activity (low vitamin K activity) of uremic HDL particles spiked with menaquinone 7 vs. that of controls was confirmed in a bioassay using human primary vascular smooth muscle cells. Kidney menaquinone 4 tissue levels were reduced in 5/6-nephrectomized versus sham-operated C57BL/6 mice after four weeks of a vitamin K rich diet. From the analyzed enzymes involved in vitamin K metabolism, kidney HMG-CoA reductase protein was reduced in both rats and patients with CKD. In a trial on the efficacy and safety of atorvastatin in 1051 patients with type 2 diabetes receiving dialysis therapy, no pronounced vitamin K deficiency was noted. However, the highest levels of PIVKA-II (biomarker of subclinical vitamin K deficiency) were noted when a statin was combined with a proton pump inhibitor. Thus, profound disturbances in lipoprotein mediated vitamin K transport and metabolism in uremia suggest that menaquinone 7 supplementation to patients on dialysis therapy has reduced efficacy.
Collapse
Affiliation(s)
- Nadine Kaesler
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany.
| | - Felix Schreibing
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany
| | - Thimoteus Speer
- Department of Internal Medicine, Nephrology and Hypertension and Cardio-Renal Medicine, Saarland University Medical Centre, Homburg, Saar, Germany; Translational Cardio-Renal Medicine, Saarland University, Homburg/Saar, Germany
| | | | - Nikolas Rapp
- Department of Biochemistry, Cardiovascular Research School Maastricht, Maastricht University, Maastricht, the Netherlands
| | | | - Nazanin Kabgani
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany
| | - Christoph Kuppe
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Vera Jankowski
- Institute of Molecular and Cardiovascular Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Leon Schurgers
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany; Department of Biochemistry, Cardiovascular Research School Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany; Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jürgen Floege
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
38
|
Zheng A, Li H, Feng Z, Liu J. Integrative Analyses Reveal Tstd1 as a Potential Modulator of HDL Cholesterol and Mitochondrial Function in Mice. Cells 2021; 10:2976. [PMID: 34831199 PMCID: PMC8616306 DOI: 10.3390/cells10112976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
High-density lipoprotein (HDL) cholesterol levels are closely associated with human health and diseases. To identify genes modulating plasma HDL levels, we integrated HDL measurements and multi-omics data collected from diverse mouse cohorts and combined a list of systems genetics methods, including quantitative trait loci (QTL) mapping analysis, mediation analysis, transcriptome-wide association analysis (TWAS), and correlation analysis. We confirmed a significant and conserved QTL for plasma HDL on chromosome 1 and identified that Tstd1 liver transcript correlates with plasma HDL in several independent mouse cohorts, suggesting Tstd1 may be a potential modulator of plasma HDL levels. Correlation analysis using over 70 transcriptomics datasets in humans and mice revealed consistent correlations between Tstd1 and genes known to be involved in cholesterol and HDL regulation. Consistent with strong enrichment in gene sets related to cholesterol and lipoproteins in the liver, mouse strains with high Tstd1 exhibited higher plasma levels of HDL, total cholesterol and other lipid markers. GeneBridge using large-scale expression datasets identified conserved and positive associations between TSTD1/Tstd1 and mitochondrial pathways, as well as cholesterol and lipid pathways in human, mouse and rat. In summary, we identified Tstd1 as a new modulator of plasma HDL and mitochondrial function through integrative systems analyses, and proposed a new mechanism of HDL modulation and a potential therapeutic target for relevant diseases. This study highlights the value of such integrative approaches in revealing molecular mechanisms of complex traits or diseases.
Collapse
Affiliation(s)
- Adi Zheng
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, 1005 Lausanne, Switzerland;
| | - Hao Li
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Zhihui Feng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China;
- University of Health and Rehabilitation Sciences, Qingdao 266071, China
| |
Collapse
|
39
|
Animal Models of Neointimal Hyperplasia and Restenosis: Species-Specific Differences and Implications for Translational Research. JACC Basic Transl Sci 2021; 6:900-917. [PMID: 34869956 PMCID: PMC8617545 DOI: 10.1016/j.jacbts.2021.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 12/29/2022]
Abstract
Neointimal hyperplasia is the major factor contributing to restenosis after angioplasty procedures. Multiple animal models exist to study basic and translational aspects of restenosis formation. Animal models differ substantially, and species-specific differences have major impact on the pathophysiology of the model. Genetic, dietary, and mechanical interventions determine the translational potential of the animal model used and have to be considered when choosing the model.
The process of restenosis is based on the interplay of various mechanical and biological processes triggered by angioplasty-induced vascular trauma. Early arterial recoil, negative vascular remodeling, and neointimal formation therefore limit the long-term patency of interventional recanalization procedures. The most serious of these processes is neointimal hyperplasia, which can be traced back to 4 main mechanisms: endothelial damage and activation; monocyte accumulation in the subintimal space; fibroblast migration; and the transformation of vascular smooth muscle cells. A wide variety of animal models exists to investigate the underlying pathophysiology. Although mouse models, with their ease of genetic manipulation, enable cell- and molecular-focused fundamental research, and rats provide the opportunity to use stent and balloon models with high throughput, both rodents lack a lipid metabolism comparable to humans. Rabbits instead build a bridge to close the gap between basic and clinical research due to their human-like lipid metabolism, as well as their size being accessible for clinical angioplasty procedures. Every different combination of animal, dietary, and injury model has various advantages and disadvantages, and the decision for a proper model requires awareness of species-specific biological properties reaching from vessel morphology to distinct cellular and molecular features.
Collapse
Key Words
- Apo, apolipoprotein
- CETP, cholesteryl ester transferase protein
- ECM, extracellular matrix
- FGF, fibroblast growth factor
- HDL, high-density lipoprotein
- LDL, low-density lipoprotein
- LDLr, LDL receptor
- PDGF, platelet-derived growth factor
- TGF, transforming growth factor
- VLDL, very low-density lipoprotein
- VSMC, vascular smooth muscle cell
- angioplasty
- animal model
- neointimal hyperplasia
- restenosis
Collapse
|
40
|
Larsen LE, van den Boogert MAW, Rios-Ocampo WA, Jansen JC, Conlon D, Chong PLE, Levels JHM, Eilers RE, Sachdev VV, Zelcer N, Raabe T, He M, Hand NJ, Drenth JPH, Rader DJ, Stroes ESG, Lefeber DJ, Jonker JW, Holleboom AG. Defective Lipid Droplet-Lysosome Interaction Causes Fatty Liver Disease as Evidenced by Human Mutations in TMEM199 and CCDC115. Cell Mol Gastroenterol Hepatol 2021; 13:583-597. [PMID: 34626841 PMCID: PMC8688563 DOI: 10.1016/j.jcmgh.2021.09.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Recently, novel inborn errors of metabolism were identified because of mutations in V-ATPase assembly factors TMEM199 and CCDC115. Patients are characterized by generalized protein glycosylation defects, hypercholesterolemia, and fatty liver disease. Here, we set out to characterize the lipid and fatty liver phenotype in human plasma, cell models, and a mouse model. METHODS AND RESULTS Patients with TMEM199 and CCDC115 mutations displayed hyperlipidemia, characterized by increased levels of lipoproteins in the very low density lipoprotein range. HepG2 hepatoma cells, in which the expression of TMEM199 and CCDC115 was silenced, and induced pluripotent stem cell (iPSC)-derived hepatocyte-like cells from patients with TMEM199 mutations showed markedly increased secretion of apolipoprotein B (apoB) compared with controls. A mouse model for TMEM199 deficiency with a CRISPR/Cas9-mediated knock-in of the human A7E mutation had marked hepatic steatosis on chow diet. Plasma N-glycans were hypogalactosylated, consistent with the patient phenotype, but no clear plasma lipid abnormalities were observed in the mouse model. In the siTMEM199 and siCCDC115 HepG2 hepatocyte models, increased numbers and size of lipid droplets were observed, including abnormally large lipid droplets, which colocalized with lysosomes. Excessive de novo lipogenesis, failing oxidative capacity, and elevated lipid uptake were not observed. Further investigation of lysosomal function revealed impaired acidification combined with impaired autophagic capacity. CONCLUSIONS Our data suggest that the hypercholesterolemia in TMEM199 and CCDC115 deficiency is due to increased secretion of apoB-containing particles. This may in turn be secondary to the hepatic steatosis observed in these patients as well as in the mouse model. Mechanistically, we observed impaired lysosomal function characterized by reduced acidification, autophagy, and increased lysosomal lipid accumulation. These findings could explain the hepatic steatosis seen in patients and highlight the importance of lipophagy in fatty liver disease. Because this pathway remains understudied and its regulation is largely untargeted, further exploration of this pathway may offer novel strategies for therapeutic interventions to reduce lipotoxicity in fatty liver disease.
Collapse
Affiliation(s)
- Lars E Larsen
- Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands; Department of Pediatrics, Section Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, The Netherlands
| | | | - Wilson A Rios-Ocampo
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Jos C Jansen
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Donna Conlon
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patrick L E Chong
- Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - J Han M Levels
- Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Roos E Eilers
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Vinay V Sachdev
- Department of Medical Biochemistry, Amsterdam University Medical Centers, location AMC, Amsterdam, The Netherlands
| | - Noam Zelcer
- Department of Medical Biochemistry, Amsterdam University Medical Centers, location AMC, Amsterdam, The Netherlands
| | - Tobias Raabe
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Miao He
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nicholas J Hand
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David J Rader
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Eric S G Stroes
- Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Dirk J Lefeber
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, Section Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Adriaan G Holleboom
- Department of Vascular Medicine, Amsterdam UMC, location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Nyandwi JB, Ko YS, Jin H, Yun SP, Park SW, Kim HJ. Rosmarinic Acid Exhibits a Lipid-Lowering Effect by Modulating the Expression of Reverse Cholesterol Transporters and Lipid Metabolism in High-Fat Diet-Fed Mice. Biomolecules 2021; 11:1470. [PMID: 34680102 PMCID: PMC8533102 DOI: 10.3390/biom11101470] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022] Open
Abstract
Hyperlipidemia is a potent risk factor for the development of cardiovascular diseases. The reverse cholesterol transport (RCT) process has been shown to alleviate hyperlipidemia and protect against cardiovascular diseases. Recently, rosmarinic acid was reported to exhibit lipid-lowering effects. However, the underlying mechanism is still unclear. This study aims to investigate whether rosmarinic acid lowers lipids by modulating the RCT process in high-fat diet (HFD)-induced hyperlipidemic C57BL/6J mice. Our results indicated that rosmarinic acid treatment significantly decreased body weight, blood glucose, and plasma total cholesterol and triglyceride levels in HFD-fed mice. Rosmarinic acid increased the expression levels of cholesterol uptake-associated receptors in liver tissues, including scavenger receptor B type 1 (SR-B1) and low-density lipoprotein receptor (LDL-R). Furthermore, rosmarinic acid treatment notably increased the expression of cholesterol excretion molecules, ATP-binding cassette G5 (ABCG5) and G8 (ABCG8) transporters, and cholesterol 7 alpha-hydroxylase A1 (CYP7A1) as well as markedly reduced cholesterol and triglyceride levels in liver tissues. In addition, rosmarinic acid facilitated fatty acid oxidation through AMP-activated protein kinase (AMPK)-mediated carnitine palmitoyltransferase 1A (CPT1A) induction. In conclusion, rosmarinic acid exhibited a lipid-lowering effect by modulating the expression of RCT-related proteins and lipid metabolism-associated molecules, confirming its potential for the prevention or treatment of hyperlipidemia-derived diseases.
Collapse
Affiliation(s)
- Jean Baptiste Nyandwi
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Kigali 4285, Rwanda
| | - Young Shin Ko
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Hana Jin
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
| |
Collapse
|
42
|
Giroud M, Jodeleit H, Prentice KJ, Bartelt A. Adipocyte function and the development of cardiometabolic disease. J Physiol 2021; 600:1189-1208. [PMID: 34555180 DOI: 10.1113/jp281979] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/31/2021] [Indexed: 11/08/2022] Open
Abstract
Obesity is a medical disorder caused by multiple mechanisms of dysregulated energy balance. A major consequence of obesity is an increased risk to develop diabetes, diabetic complications and cardiovascular disease. While a better understanding of the molecular mechanisms linking obesity, insulin resistance and cardiovascular disease is needed, translational research of the human pathology is hampered by the available cellular and rodent model systems. Major barriers are the species-specific differences in energy balance, vascular biology and adipose tissue physiology, especially related to white and brown adipocytes, and adipose tissue browning. In rodents, non-shivering thermogenesis is responsible for a large part of energy expenditure, but humans possess much less thermogenic fat, which means temperature is an important variable in translational research. Mouse models with predisposition to dyslipidaemia housed at thermoneutrality and fed a high-fat diet more closely reflect human physiology. Also, adipocytes play a key role in the endocrine regulation of cardiovascular function. Adipocytes secrete a variety of hormones, lipid mediators and other metabolites that directly influence the local microenvironment as well as distant tissues. This is specifically apparent in perivascular depots, where adipocytes modulate vascular function and inflammation. Altogether, these mechanisms highlight the critical role of adipocytes in the development of cardiometabolic disease.
Collapse
Affiliation(s)
- Maude Giroud
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany
| | - Henrika Jodeleit
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Bavaria, Germany
| | - Kacey J Prentice
- Department of Molecular Metabolism & Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Bavaria, Germany.,Department of Molecular Metabolism & Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
43
|
Estévez-Vázquez O, Benedé-Ubieto R, Guo F, Gómez-Santos B, Aspichueta P, Reissing J, Bruns T, Sanz-García C, Sydor S, Bechmann LP, Maranillo E, Sañudo JR, Vázquez MT, Lamas-Paz A, Morán L, Mazariegos MS, Ciudin A, Pericàs JM, Peligros MI, Vaquero J, Martínez-Naves E, Liedtke C, Regueiro JR, Trautwein C, Bañares R, Cubero FJ, Nevzorova YA. Fat: Quality, or Quantity? What Matters Most for the Progression of Metabolic Associated Fatty Liver Disease (MAFLD). Biomedicines 2021; 9:biomedicines9101289. [PMID: 34680405 PMCID: PMC8533605 DOI: 10.3390/biomedicines9101289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/13/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023] Open
Abstract
Objectives: Lately, many countries have restricted or even banned transfat, and palm oil has become a preferred replacement for food manufacturers. Whether palm oil is potentially an unhealthy food mainly due to its high content of saturated Palmitic Acid (PA) is a matter of debate. The aim of this study was to test whether qualitative aspects of diet such as levels of PA and the fat source are risk factors for Metabolic Syndrome (MS) and Metabolic Associated Fatty Liver Disease (MAFLD). Methods: C57BL/6 male mice were fed for 14 weeks with three types of Western diet (WD): 1. LP-WD—low concentration of PA (main fat source—corn and soybean oils); 2. HP-WD—high concentration of PA (main fat source—palm oil); 3. HP-Trans-WD—high concentration of PA (mainly transfat). Results: All types of WD caused weight gain, adipocyte enlargement, hepatomegaly, lipid metabolism alterations, and steatohepatitis. Feeding with HP diets led to more prominent obesity, hypercholesterolemia, stronger hepatic injury, and fibrosis. Only the feeding with HP-Trans-WD resulted in glucose intolerance and elevation of serum transaminases. Brief withdrawal of WDs reversed MS and signs of MAFLD. However, mild hepatic inflammation was still detectable in HP groups. Conclusions: HP and HP-Trans-WD play a crucial role in the genesis of MS and MAFLD.
Collapse
Affiliation(s)
- Olga Estévez-Vázquez
- Department of Physiology, Genetics and Microbiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; (O.E.-V.); (R.B.-U.)
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
| | - Raquel Benedé-Ubieto
- Department of Physiology, Genetics and Microbiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; (O.E.-V.); (R.B.-U.)
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
| | - Feifei Guo
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
| | - Beatriz Gómez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, 48940 Leioa, Spain; (B.G.-S.); (P.A.)
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, 48940 Leioa, Spain; (B.G.-S.); (P.A.)
- Biocruces Health Research Institute, 48903 Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28220 Madrid, Spain; (J.M.P.); (J.V.)
| | - Johanna Reissing
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (C.L.); (C.T.)
| | - Tony Bruns
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (C.L.); (C.T.)
| | - Carlos Sanz-García
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
| | - Svenja Sydor
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, 44801 Bochum, Germany; (S.S.); (L.P.B.)
| | - Lars P. Bechmann
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, 44801 Bochum, Germany; (S.S.); (L.P.B.)
| | - Eva Maranillo
- Department of Human Anatomy and Embryology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (E.M.); (J.R.S.); (M.T.V.)
| | - José Ramón Sañudo
- Department of Human Anatomy and Embryology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (E.M.); (J.R.S.); (M.T.V.)
| | - María Teresa Vázquez
- Department of Human Anatomy and Embryology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (E.M.); (J.R.S.); (M.T.V.)
| | - Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
| | - Laura Morán
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
| | - Marina S. Mazariegos
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
| | - Andreea Ciudin
- Endocrinology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute for Research (VHIR), 08035 Barcelona, Spain;
| | - Juan M. Pericàs
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28220 Madrid, Spain; (J.M.P.); (J.V.)
- Liver Unit, Internal Medicine Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute for Research (VHIR), 08035 Barcelona, Spain
| | - María Isabel Peligros
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain;
| | - Javier Vaquero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28220 Madrid, Spain; (J.M.P.); (J.V.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
- 12 de Octubre Health Research Institute (imas12), 28041 Madrid, Spain
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (C.L.); (C.T.)
| | - José R. Regueiro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
- 12 de Octubre Health Research Institute (imas12), 28041 Madrid, Spain
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (C.L.); (C.T.)
| | - Rafael Bañares
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28220 Madrid, Spain; (J.M.P.); (J.V.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28220 Madrid, Spain; (J.M.P.); (J.V.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
| | - Yulia A. Nevzorova
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (F.G.); (C.S.-G.); (A.L.-P.); (L.M.); (M.S.M.); (E.M.-N.); (J.R.R.); (R.B.); (F.J.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28220 Madrid, Spain; (J.M.P.); (J.V.)
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (C.L.); (C.T.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
- Correspondence: ; Tel.: +49-(0)241-80-80662; Fax: +49-(0)241-80-82455
| |
Collapse
|
44
|
Ceballos-Gutiérrez A, Rodríguez-Hernández A, Álvarez-Valadez MDR, Limón-Miranda S, Andrade F, Figueroa-Gutiérrez A, Díaz-Reval I, Apolinar-Iribe A, Castro-Sánchez L, Alamilla J, Sánchez-Pastor E, Virgen-Ortiz A. ZnO Nanoparticles Induce Dyslipidemia and Atherosclerotic Lesions Leading to Changes in Vascular Contractility and Cannabinoid Receptors Expression as Well as Increased Blood Pressure. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2319. [PMID: 34578635 PMCID: PMC8472382 DOI: 10.3390/nano11092319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 12/04/2022]
Abstract
ZnO nanoparticles (ZnONPs) have been shown to have therapeutic potential in some diseases such as diabetes and cancer. However, concentration-dependent adverse effects have also been reported. Studies which evaluate the effects of ZnONPs on the cardiovascular system are scarce. This study aimed to evaluate the cardiovascular effects of a low dose of ZnONPs administered chronically in healthy rats. Changes in dyslipidemia biomarkers, blood pressure, aortic wall structure, vascular contractility, and expression of cannabinoid receptors in the aorta wall were evaluated. Healthy rats were divided into two groups: control or treated (one, two, and three months). The treated rats received an oral dose of 10 mg/kg/day. The results showed that treatment with ZnONPs induced dyslipidemia from the first month, increasing atherosclerosis risk, which was confirmed by presence of atherosclerotic alterations revealed by aorta histological analysis. In in vitro assays, ZnONPs modified the aorta contractile activity in response to the activation of cannabinoid receptors (CB1 and CB2). The expression of CB1 and CB2 was modified as well. Moreover, ZnONPs elicited an increase in blood pressure. In conclusion, long-time oral administration of ZnONPs induce dyslipidemia and atherosclerosis eliciting alterations in aorta contractility, CB1 and CB2 receptors expression, and an increase in blood pressure in healthy rats.
Collapse
Affiliation(s)
| | | | | | - Saraí Limón-Miranda
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Unidad Regional Sur, Universidad de Sonora, Navojoa 85880, Mexico;
| | | | | | - Irene Díaz-Reval
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Mexico; (M.d.R.Á.-V.); (I.D.-R.)
| | | | - Luis Castro-Sánchez
- Centro Universitario de Investigaciones Biomédicas, CONACYT-Universidad de Colima, Universidad de Colima, Colima 28045, Mexico; (L.C.-S.); (J.A.)
| | - Javier Alamilla
- Centro Universitario de Investigaciones Biomédicas, CONACYT-Universidad de Colima, Universidad de Colima, Colima 28045, Mexico; (L.C.-S.); (J.A.)
| | - Enrique Sánchez-Pastor
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Mexico; (M.d.R.Á.-V.); (I.D.-R.)
| | - Adolfo Virgen-Ortiz
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Mexico; (M.d.R.Á.-V.); (I.D.-R.)
| |
Collapse
|
45
|
Eng JM, Estall JL. Diet-Induced Models of Non-Alcoholic Fatty Liver Disease: Food for Thought on Sugar, Fat, and Cholesterol. Cells 2021; 10:cells10071805. [PMID: 34359974 PMCID: PMC8303413 DOI: 10.3390/cells10071805] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects approximately 1 in 4 people worldwide and is a major burden to health care systems. A major concern in NAFLD research is lack of confidence in pre-clinical animal models, raising questions regarding translation to humans. Recently, there has been renewed interest in creating dietary models of NAFLD with higher similarity to human diets in hopes to better recapitulate disease pathology. This review summarizes recent research comparing individual roles of major dietary components to NAFLD and addresses common misconceptions surrounding frequently used diet-based NAFLD models. We discuss the effects of glucose, fructose, and sucrose on the liver, and how solid vs. liquid sugar differ in promoting disease. We consider studies on dosages of fat and cholesterol needed to promote NAFLD versus NASH, and discuss important considerations when choosing control diets, mouse strains, and diet duration. Lastly, we provide our recommendations on amount and type of sugar, fat, and cholesterol to include when modelling diet-induced NAFLD/NASH in mice.
Collapse
Affiliation(s)
- James M. Eng
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada;
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Jennifer L. Estall
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada;
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Correspondence: ; Tel.: +1-(514)-987-5688
| |
Collapse
|
46
|
Mohammed Y, Michaud SA, Pětrošová H, Yang J, Ganguly M, Schibli D, Flenniken AM, Nutter LMJ, Adissu HA, Lloyd KCK, McKerlie C, Borchers CH. Proteotyping of knockout mouse strains reveals sex- and strain-specific signatures in blood plasma. NPJ Syst Biol Appl 2021; 7:25. [PMID: 34050187 PMCID: PMC8163790 DOI: 10.1038/s41540-021-00184-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 04/25/2021] [Indexed: 11/24/2022] Open
Abstract
We proteotyped blood plasma from 30 mouse knockout strains and corresponding wild-type mice from the International Mouse Phenotyping Consortium. We used targeted proteomics with internal standards to quantify 375 proteins in 218 samples. Our results provide insights into the manifested effects of each gene knockout at the plasma proteome level. We first investigated possible contamination by erythrocytes during sample preparation and labeled, in one case, up to 11 differential proteins as erythrocyte originated. Second, we showed that differences in baseline protein abundance between female and male mice were evident in all mice, emphasizing the necessity to include both sexes in basic research, target discovery, and preclinical effect and safety studies. Next, we identified the protein signature of each gene knockout and performed functional analyses for all knockout strains. Further, to demonstrate how proteome analysis identifies the effect of gene deficiency beyond traditional phenotyping tests, we provide in-depth analysis of two strains, C8a-/- and Npc2+/-. The proteins encoded by these genes are well-characterized providing good validation of our method in homozygous and heterozygous knockout mice. Ig alpha chain C region, a poorly characterized protein, was among the differentiating proteins in C8a-/-. In Npc2+/- mice, where histopathology and traditional tests failed to differentiate heterozygous from wild-type mice, our data showed significant difference in various lysosomal storage disease-related proteins. Our results demonstrate how to combine absolute quantitative proteomics with mouse gene knockout strategies to systematically study the effect of protein absence. The approach used here for blood plasma is applicable to all tissue protein extracts.
Collapse
Affiliation(s)
- Yassene Mohammed
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada.
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands.
| | - Sarah A Michaud
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada.
| | - Helena Pětrošová
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Juncong Yang
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Milan Ganguly
- The Center for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - David Schibli
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Ann M Flenniken
- The Center for Phenogenomics, Toronto, ON, Canada
- Sinai Health Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Lauryl M J Nutter
- The Center for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | | | - K C Kent Lloyd
- Department of Surgery, School of Medicine, and Mouse Biology Program, University of California, Davis, CA, USA
| | | | - Christoph H Borchers
- Proteomics Centre, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC, Canada.
- Department of Data Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Skolkovo Innovation Center, Moscow, Russia.
| |
Collapse
|
47
|
Development of a novel, sensitive translational immunoassay to detect plasma glial fibrillary acidic protein (GFAP) after murine traumatic brain injury. ALZHEIMERS RESEARCH & THERAPY 2021; 13:58. [PMID: 33678186 PMCID: PMC7938597 DOI: 10.1186/s13195-021-00793-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/17/2021] [Indexed: 12/21/2022]
Abstract
Background Glial fibrillary acidic protein (GFAP) has emerged as a promising fluid biomarker for several neurological indications including traumatic brain injury (TBI), a leading cause of death and disability worldwide. In humans, serum or plasma GFAP levels can predict brain abnormalities including hemorrhage on computed tomography (CT) scans and magnetic resonance imaging (MRI). However, assays to quantify plasma or serum GFAP in preclinical models are not yet available. Methods We developed and validated a novel sensitive GFAP immunoassay assay for mouse plasma on the Meso Scale Discovery immunoassay platform and validated assay performance for robustness, precision, limits of quantification, dilutional linearity, parallelism, recovery, stability, selectivity, and pre-analytical factors. To provide proof-of-concept data for this assay as a translational research tool for TBI and Alzheimer’s disease (AD), plasma GFAP was measured in mice exposed to TBI using the Closed Head Impact Model of Engineered Rotational Acceleration (CHIMERA) model and in APP/PS1 mice with normal or reduced levels of plasma high-density lipoprotein (HDL). Results We performed a partial validation of our novel assay and found its performance by the parameters studied was similar to assays used to quantify human GFAP in clinical neurotrauma blood specimens and to assays used to measure murine GFAP in tissues. Specifically, we demonstrated an intra-assay CV of 5.0%, an inter-assay CV of 7.2%, a lower limit of detection (LLOD) of 9.0 pg/mL, a lower limit of quantification (LLOQ) of 24.8 pg/mL, an upper limit of quantification (ULOQ) of at least 16,533.9 pg/mL, dilution linearity of calibrators from 20 to 200,000 pg/mL with 90–123% recovery, dilution linearity of plasma specimens up to 32-fold with 96–112% recovery, spike recovery of 67–100%, and excellent analyte stability in specimens exposed to up to 7 freeze-thaw cycles, 168 h at 4 °C, 24 h at room temperature (RT), or 30 days at − 20 °C. We also observed elevated plasma GFAP in mice 6 h after TBI and in aged APP/PS1 mice with plasma HDL deficiency. This assay also detects GFAP in serum. Conclusions This novel assay is a valuable translational tool that may help to provide insights into the mechanistic pathophysiology of TBI and AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00793-9.
Collapse
|
48
|
Gan ES, Tan HC, Le DHT, Huynh TT, Wills B, Seidah NG, Ooi EE, Yacoub S. Dengue virus induces PCSK9 expression to alter antiviral responses and disease outcomes. J Clin Invest 2021; 130:5223-5234. [PMID: 32644974 PMCID: PMC7524462 DOI: 10.1172/jci137536] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022] Open
Abstract
Dengue virus (DENV) infection requires cholesterol as a proviral factor, although statin treatment did not show antiviral efficacy in patients with dengue. Here, we show that DENV infection manipulated cholesterol metabolism in cells residing in low-oxygen microenvironments (hypoxia) such as in the liver, spleen, and lymph nodes. DENV infection induced expression of proprotein convertase subtilisin/kexin type 9 (PCSK9), which reduces low-density lipoprotein receptor (LDLR) recycling and hence cholesterol uptake. We found that, whereas LDLR uptake would have distributed cholesterol throughout the various cell compartments, de novo cholesterol synthesis enriched this lipid in the endoplasmic reticulum (ER). With cholesterol enrichment in the ER, ER-resident STING and type I IFN (IFN) activation was repressed during DENV infection. Our in vitro findings were further supported by the detection of elevated plasma PCSK9 levels in patients with dengue with high viremia and increased severity of plasma leakage. Our findings therefore suggest that PCSK9 plays a hitherto unrecognized role in dengue pathogenesis and that PCSK9 inhibitors could be a suitable host-directed treatment for patients with dengue.
Collapse
Affiliation(s)
| | - Hwee Cheng Tan
- Duke-National University of Singapore Medical School, Singapore
| | - Duyen Huynh Thi Le
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam
| | - Trieu Trung Huynh
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam
| | - Bridget Wills
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute, Université de Montréal, Montréal, Québec, Canada
| | - Eng Eong Ooi
- Duke-National University of Singapore Medical School, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore.,SingHealth Duke-National University of Singapore Global Health Institute, Singapore.,Antimicrobial Resistance Interdisciplinary Research Group, Singapore MIT Alliance in Research and Technology, Singapore
| | - Sophie Yacoub
- Oxford University Clinical Research Unit (OUCRU), Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom.,Antimicrobial Resistance Interdisciplinary Research Group, Singapore MIT Alliance in Research and Technology, Singapore
| |
Collapse
|
49
|
Trinder M, Wang Y, Madsen CM, Ponomarev T, Bohunek L, Daisely BA, Julia Kong H, Blauw LL, Nordestgaard BG, Tybjærg-Hansen A, Wurfel MM, Russell JA, Walley KR, Rensen PCN, Boyd JH, Brunham LR. Inhibition of Cholesteryl Ester Transfer Protein Preserves High-Density Lipoprotein Cholesterol and Improves Survival in Sepsis. Circulation 2020; 143:921-934. [PMID: 33228395 DOI: 10.1161/circulationaha.120.048568] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The high-density lipoprotein hypothesis of atherosclerosis has been challenged by clinical trials of cholesteryl ester transfer protein (CETP) inhibitors, which failed to show significant reductions in cardiovascular events. Plasma levels of high-density lipoprotein cholesterol (HDL-C) decline drastically during sepsis, and this phenomenon is explained, in part, by the activity of CETP, a major determinant of plasma HDL-C levels. We tested the hypothesis that genetic or pharmacological inhibition of CETP would preserve high-density lipoprotein levels and decrease mortality in clinical cohorts and animal models of sepsis. METHODS We examined the effect of a gain-of-function variant in CETP (rs1800777, p.Arg468Gln) and a genetic score for decreased CETP function on 28-day sepsis survival using Cox proportional hazard models adjusted for age and sex in the UK Biobank (n=5949), iSPAAR (Identification of SNPs Predisposing to Altered Acute Lung Injury Risk; n=882), Copenhagen General Population Study (n=2068), Copenhagen City Heart Study (n=493), Early Infection (n=200), St Paul's Intensive Care Unit 2 (n=203), and Vasopressin Versus Norepinephrine Infusion in Patients With Septic Shock studies (n=632). We then studied the effect of the CETP inhibitor, anacetrapib, in adult female APOE*3-Leiden mice with or without human CETP expression using the cecal-ligation and puncture model of sepsis. RESULTS A fixed-effect meta-analysis of all 7 cohorts found that the CETP gain-of-function variant was significantly associated with increased risk of acute sepsis mortality (hazard ratio, 1.44 [95% CI, 1.22-1.70]; P<0.0001). In addition, a genetic score for decreased CETP function was associated with significantly decreased sepsis mortality in the UK Biobank (hazard ratio, 0.77 [95% CI, 0.59-1.00] per 1 mmol/L increase in HDL-C) and iSPAAR cohorts (hazard ratio, 0.60 [95% CI, 0.37-0.98] per 1 mmol/L increase in HDL-C). APOE*3-Leiden.CETP mice treated with anacetrapib had preserved levels of HDL-C and apolipoprotein-AI and increased survival relative to placebo treatment (70.6% versus 35.3%, Log-rank P=0.03), whereas there was no effect of anacetrapib on the survival of APOE*3-Leiden mice that did not express CETP (50.0% versus 42.9%, Log-rank P=0.87). CONCLUSIONS Clinical genetics and humanized mouse models suggest that inhibiting CETP may preserve high-density lipoprotein levels and improve outcomes for individuals with sepsis.
Collapse
Affiliation(s)
- Mark Trinder
- Centre for Heart Lung Innovation (M.T., T.P., L.B., H.J.K., J.A.R., K.R.W., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada.,Experimental Medicine Program (M.T., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada
| | - Yanan Wang
- Department of Medicine, Division of Endocrinology (Y.W., L.L.B., P.C.N.R.), Leiden University Medical Center, The Netherlands
| | - Christian M Madsen
- Department of Clinical Biochemistry (C.M.M., B.G.N., J.A.R.), Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., B.G.N., A.T.-H.), Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., B.G.N., A.T.-H.)
| | - Tatjana Ponomarev
- Centre for Heart Lung Innovation (M.T., T.P., L.B., H.J.K., J.A.R., K.R.W., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada
| | | | - Brendan A Daisely
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada (B.A.D.)
| | - HyeJin Julia Kong
- Centre for Heart Lung Innovation (M.T., T.P., L.B., H.J.K., J.A.R., K.R.W., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada
| | - Lisanne L Blauw
- Department of Medicine, Division of Endocrinology (Y.W., L.L.B., P.C.N.R.), Leiden University Medical Center, The Netherlands
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry (C.M.M., B.G.N., J.A.R.), Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., B.G.N., A.T.-H.), Copenhagen University Hospital, Denmark.,The Copenhagen City Heart Study, Frederiksberg Hospital (B.G.N., A.T.-H.), Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., B.G.N., A.T.-H.)
| | - Anne Tybjærg-Hansen
- The Copenhagen General Population Study (C.M.M., B.G.N., A.T.-H.), Copenhagen University Hospital, Denmark.,Herlev Gentofte Hospital, Department of Clinical Biochemistry, Rigshospitalet (A.T.-H.), Copenhagen University Hospital, Denmark.,The Copenhagen City Heart Study, Frederiksberg Hospital (B.G.N., A.T.-H.), Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., B.G.N., A.T.-H.)
| | - Mark M Wurfel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle (M.M.W., K.R.W.)
| | - James A Russell
- Centre for Heart Lung Innovation (M.T., T.P., L.B., H.J.K., J.A.R., K.R.W., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada.,Department of Clinical Biochemistry (C.M.M., B.G.N., J.A.R.), Copenhagen University Hospital, Denmark
| | - Keith R Walley
- Centre for Heart Lung Innovation (M.T., T.P., L.B., H.J.K., J.A.R., K.R.W., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle (M.M.W., K.R.W.)
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology (Y.W., L.L.B., P.C.N.R.), Leiden University Medical Center, The Netherlands
| | - John H Boyd
- Centre for Heart Lung Innovation (M.T., T.P., L.B., H.J.K., J.A.R., K.R.W., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada.,Experimental Medicine Program (M.T., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada.,Department of Medicine (J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada
| | - Liam R Brunham
- Centre for Heart Lung Innovation (M.T., T.P., L.B., H.J.K., J.A.R., K.R.W., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada.,Experimental Medicine Program (M.T., J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada.,Department of Medicine (J.H.B., L.R.B.), University of British Columbia, Vancouver, Canada
| |
Collapse
|
50
|
Loupy KM, Lee T, Zambrano CA, Elsayed AI, D'Angelo HM, Fonken LK, Frank MG, Maier SF, Lowry CA. Alzheimer's Disease: Protective Effects of Mycobacterium vaccae, a Soil-Derived Mycobacterium with Anti-Inflammatory and Anti-Tubercular Properties, on the Proteomic Profiles of Plasma and Cerebrospinal Fluid in Rats. J Alzheimers Dis 2020; 78:965-987. [PMID: 33074227 DOI: 10.3233/jad-200568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is an inflammatory neurodegenerative disease that may be associated with prior bacterial infections. Microbial "old friends" can suppress exaggerated inflammation in response to disease-causing infections or increase clearance of pathogens such as Mycobacterium tuberculosis, which causes tuberculosis (TB). One such "old friend" is Mycobacterium vaccae NCTC 11659, a soil-derived bacterium that has been proposed either as a vaccine for prevention of TB, or as immunotherapy for the treatment of TB when used alongside first line anti-TB drug treatment. OBJECTIVE The goal of this study was to use a hypothesis generating approach to explore the effects of M. vaccae on physiological changes in the plasma and cerebrospinal fluid (CSF). METHODS Liquid chromatography-tandem mass spectrometry-based proteomics were performed in plasma and CSF of adult male rats after immunization with a heat-killed preparation of M. vaccae NCTC 11659 or borate-buffered saline vehicle. Gene enrichment analysis and analysis of protein-protein interactions were performed to integrate physiological network changes in plasma and CSF. We used RT-qPCR to assess immune and metabolic gene expression changes in the hippocampus. RESULTS In both plasma and CSF, immunization with M. vaccae increased proteins associated with immune activation and downregulated proteins corresponding to lipid (including phospholipid and cholesterol) metabolism. Immunization with M. vaccae also increased hippocampal expression of interleukin-4 (IL-4) mRNA, implicating anti-inflammatory effects in the central nervous system. CONCLUSION M. vaccae alters host immune activity and lipid metabolism. These data are consistent with the hypothesis that microbe-host interactions may protect against possible infection-induced, inflammation-related cognitive impairments.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Thomas Lee
- Central Analytical Laboratory and Mass Spectrometry Facility, Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Cristian A Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Heather M D'Angelo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Senior Fellow, inVIVO Planetary Health, of the Worldwide Universities Network (WUN), West New York, NJ, USA
| |
Collapse
|