1
|
Frisk C, Ekström M, Eriksson MJ, Corbascio M, Hage C, Persson H, Linde C, Persson B. Characteristics of gene expression in epicardial adipose tissue and subcutaneous adipose tissue in patients at risk for heart failure undergoing coronary artery bypass grafting. BMC Genomics 2024; 25:938. [PMID: 39375631 PMCID: PMC11457432 DOI: 10.1186/s12864-024-10851-9] [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: 08/16/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Epicardial adipose tissue (EAT) surrounds the heart and is hypothesised to play a role in the development of heart failure (HF). In this study, we first investigated the differences in gene expression between epicardial adipose tissue (EAT) and subcutaneous adipose tissue (SAT) in patients undergoing elective coronary artery bypass graft (CABG) surgery (n = 21; 95% male). Secondly, we examined the association between EAT and SAT in patients at risk for HF stage A (n = 12) and in pre-HF patients, who show signs but not symptoms of HF, stage B (n = 9). RESULTS The study confirmed a distinct separation between EAT and SAT. In EAT 17 clusters of genes were present, of which several novel gene modules are associated with characteristics of HF. Notably, seven gene modules showed significant correlation to measures of HF, such as end diastolic left ventricular posterior wall thickness, e'mean, deceleration time and BMI. One module was particularly distinct in EAT when compared to SAT, featuring key genes such as FLT4, SEMA3A, and PTX3, which are implicated in angiogenesis, inflammation regulation, and tissue repair, suggesting a unique role in EAT linked to left ventricular dysfunction. Genetic expression was compared in EAT across all pre-HF and normal phenotypes, revealing small genetic changes in the form of 18 differentially expressed genes in ACC/AHA Stage A vs. Stage B. CONCLUSIONS The roles of subcutaneous and epicardial fat are clearly different. We highlight the gene expression difference in search of potential modifiers of HF progress. The true implications of our findings should be corroborated in other studies since HF ACC/AHA stage B patients are common and carry a considerable risk for progression to symptomatic HF.
Collapse
Affiliation(s)
- Christoffer Frisk
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, Uppsala, S-751 24, Sweden
| | - Mattias Ekström
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, S-182 88, Sweden
- Department of Cardiology, Danderyd Hospital, Stockholm, S-182 88, Sweden
| | - Maria J Eriksson
- Department of Clinical Physiology, Karolinska University Hospital, Stockholm, S-171 76, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, S-171 77, Sweden
| | - Matthias Corbascio
- Department of Clinical Physiology, Karolinska University Hospital, Stockholm, S-171 76, Sweden
- Department of Thoracic Surgery, Karolinska University Hospital, Stockholm, S-171 76, Sweden
| | - Camilla Hage
- Department of Medicine, Karolinska Institutet, Stockholm, S-171 77, Sweden
- Karolinska University Hospital, Heart and Vascular Theme, Stockholm, S-171 76, Sweden
| | - Hans Persson
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, S-182 88, Sweden
- Department of Cardiology, Danderyd Hospital, Stockholm, S-182 88, Sweden
| | - Cecilia Linde
- Department of Medicine, Karolinska Institutet, Stockholm, S-171 77, Sweden
- Karolinska University Hospital, Heart and Vascular Theme, Stockholm, S-171 76, Sweden
| | - Bengt Persson
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, Uppsala, S-751 24, Sweden.
| |
Collapse
|
2
|
Sun J, Xu J, Liu Y, Xu X, Zhang S, Hao Y, Lin Y, Han Y, Li F, Yuan H. Proteomic and metabolomic analyses reveal the novel targets of spermine for alleviating diabetic cardiomyopathy in type II diabetic mice. Front Cardiovasc Med 2022; 9:1022861. [PMID: 36312255 PMCID: PMC9614018 DOI: 10.3389/fcvm.2022.1022861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the most serious complications of diabetes. Recent cardiology studies suggest that spermine has a cardioprotective effect. Here, we used proteomic and metabolomic analyses to reveal the underlying research targets in a type II diabetic (T2D) mouse model treated with spermine. Left ventricular tissues from nine mice (Control group, three; T2D group, three; T2D+SP group, three) were excised and analyzed. Quantitative analysis of the global proteome and metabolome was performed using the 4D label-free technique and untargeted metabolomics, respectively, and differentially expressed proteins (DEPs) and metabolites were used to perform bioinformatic analyses. A total of 169 DEPs were identified in T2D/Control group, including 115 upregulated and 54 downregulated proteins. Furthermore, 16 DEPs were identified in T2D+SP/T2D group, where these DEPs were found highly enriched in the cellular, metabolic processes, biological regulation, response to stimulus, and immune system process. The results of association analysis between proteomics and metabolomics showed that SP could affect the production of 51 metabolites by regulating the expression of 16 DEPs in the T2D+SP/T2D group. We also found that PRKG1 was closely related to the expressions of 10 overlapping metabolites between db/db and SP-treated mice. Our findings provide insights into the underlying mechanisms for DCM and suggest the potential applicability of utilizing spermine on protecting against DCM-associated cardiac function deterioration.
Collapse
Affiliation(s)
- Jian Sun
- School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Jiyu Xu
- School of Medical Imaging, Mudanjiang Medical University, Mudanjiang, China
| | - Yong Liu
- Research Department, Animal Research Institute, Mudanjiang Medical University, Mudanjiang, China
| | - Xiaoyi Xu
- The First Clinical School of Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Shumin Zhang
- School of Stomatology, Mudanjiang Medical University, Mudanjiang, China
| | - Yankun Hao
- School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Yitong Lin
- School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Yue Han
- School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Feiya Li
- Department of Laboratory Medicine and Pathobiology, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Hui Yuan
- School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, China,School of Stomatology, Mudanjiang Medical University, Mudanjiang, China,*Correspondence: Hui Yuan
| |
Collapse
|
3
|
Wang Q, Huang J, Liu S, Wang C, Jin Y, Lai H, Tu W. Aberrant hepatic lipid metabolism associated with gut microbiota dysbiosis triggers hepatotoxicity of novel PFOS alternatives in adult zebrafish. ENVIRONMENT INTERNATIONAL 2022; 166:107351. [PMID: 35738203 DOI: 10.1016/j.envint.2022.107351] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 05/23/2023]
Abstract
Perfluorooctane sulfonate (PFOS) has been reported to induce hepatotoxicity in wildlife and humans. Novel PFOS alternatives have been widely used following restrictions on PFOS, but little is known about their potential toxicity. Here, the first comprehensive investigation on the chronic hepatotoxicity and underlying molecular mechanisms of PFOS, 6:2Cl-PFESA (F-53B), and sodium p-perfluorous nonenoxybenzene sulfonate (OBS) was carried out on adult zebrafish through a histopathological examination, biochemical measurement, and multi-omics analysis. PFOS and its alternatives caused changes in liver histopathology and liver function indices in the order of F-53B > PFOS > OBS, which was consistent with their concentration in the liver. In silico modeling and transcriptional profiles suggested that the aberrant hepatic lipid metabolism induced by F-53B and PFOS was initiated by the action on peroxisome proliferator-activated receptor γ (PPARγ), which triggered changes in downstream genes transcription and led to an imbalance between lipid synthesis and expenditure. Gut microbiome analysis provided another novel mechanistic perspective that changes in the abundance of Legionella, Ralstonia, Brevundimonas, Alphaproteobacteria, Plesiomonas, and Hyphomicrobium might link to alterations in the PPAR pathway based on their significant correlation. This study provides insight into the molecular mechanisms of hepatotoxicity induced by PFOS and its novel alternatives and highlights the need for concern about their environmental exposure risks.
Collapse
Affiliation(s)
- Qiyu Wang
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China
| | - Jing Huang
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China; School of Land Resources and Environment, Jiangxi Agricultural University, Nanchang 330045, China
| | - Shuai Liu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China
| | - Caiyun Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| | - Hong Lai
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330012, China
| | - Wenqing Tu
- School of Land Resources and Environment, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
4
|
Gautier T, Deckert V, Nguyen M, Desrumaux C, Masson D, Lagrost L. New therapeutic horizons for plasma phospholipid transfer protein (PLTP): Targeting endotoxemia, infection and sepsis. Pharmacol Ther 2021; 236:108105. [PMID: 34974028 DOI: 10.1016/j.pharmthera.2021.108105] [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: 11/06/2021] [Revised: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022]
Abstract
Phospholipid Transfer Protein (PLTP) transfers amphiphilic lipids between circulating lipoproteins and between lipoproteins, cells and tissues. Indeed, PLTP is a major determinant of the plasma levels, turnover and functionality of the main lipoprotein classes: very low-density lipoproteins (VLDL), low-density lipoproteins (LDL) and high-density lipoproteins (HDL). To date, most attention has been focused on the role of PLTP in the context of cardiometabolic diseases, with additional insights in neurodegenerative diseases and immunity. Importantly, beyond its influence on plasma triglyceride and cholesterol transport, PLTP plays a key role in the modulation of the immune response, with immediate relevance to a wide range of inflammatory diseases including bacterial infection and sepsis. Indeed, emerging evidence supports the role of PLTP, in the context of its association with lipoproteins, in the neutralization and clearance of bacterial lipopolysaccharides (LPS) or endotoxins. LPS are amphipathic molecules originating from Gram-negative bacteria which harbor major pathogen-associated patterns, triggering an innate immune response in the host. Although the early inflammatory reaction constitutes a key step in the anti-microbial defense of the organism, it can lead to a dysregulated inflammatory response and to hemodynamic disorders, organ failure and eventually death. Moreover, and in addition to endotoxemia and acute inflammation, small amounts of LPS in the circulation can induce chronic, low-grade inflammation with long-term consequences in several metabolic disorders such as atherosclerosis, obesity and diabetes. After an updated overview of the role of PLTP in lipid transfer, lipoprotein metabolism and related diseases, current knowledge of its impact on inflammation, infection and sepsis is critically appraised. Finally, the relevance of PLTP as a new player and novel therapeutic target in the fight against inflammatory diseases is considered.
Collapse
Affiliation(s)
- Thomas Gautier
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France.
| | - Valérie Deckert
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Maxime Nguyen
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Service Anesthésie-Réanimation Chirurgicale, Dijon University Hospital, Dijon, France
| | - Catherine Desrumaux
- INSERM, U1198, Montpellier, France; Faculty of Sciences, Université Montpellier, Montpellier, France
| | - David Masson
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Plateau Automatisé de Biochimie, Dijon University Hospital, Dijon, France
| | - Laurent Lagrost
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Service de la Recherche, Dijon University Hospital, Dijon, France.
| |
Collapse
|
5
|
Association between the PLTP rs4810479 SNP and Serum Lipid Traits in the Chinese Maonan and Han Populations. Genet Res (Camb) 2021; 2021:9925272. [PMID: 34385888 PMCID: PMC8328490 DOI: 10.1155/2021/9925272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/22/2021] [Indexed: 11/23/2022] Open
Abstract
The association between the phospholipid transfer protein (PLTP) gene rs4810479 single-nucleotide polymorphism (SNP) and serum lipid levels is largely unknown. This investigation aimed to evaluate the relationship between the PLTP rs4810479 SNP, several environmental risk factors, and serum lipid parameters in the Chinese Maonan and Han nationalities. Polymerase chain reaction-restriction fragment length polymorphism, gel electrophoresis, and direct sequencing were employed to determine the PLTP rs4810479 genotypes in 633 Maonan and 646 Han participants. The frequencies of CC, CT, and TT genotypes and the C allele were different between Maonan and Han groups (29.07%, 53.08%, 17.85%, and 55.61% vs. 35.60%, 49.70%, 14.70%, and 60.45%, respectively, P < 0.05). The C allele carriers in the Maonan group had higher high-density lipoprotein cholesterol levels than the C allele noncarriers, but this finding was only found in Maonan males but not in females. The C allele carriers in Han males had lower total cholesterol and low-density lipoprotein cholesterol levels than the C allele noncarriers. Serum lipid profiles were also affected by several traditional cardiovascular risk factors in both populations. There might be an ethnic- and/or sex-specific association between the PLTP rs4810479 SNP and serum lipid traits.
Collapse
|
6
|
Wang H, Chen MH, Chen W, Zhang JG, Qin SC. Roles and mechanisms of phospholipid transfer protein in the development of Alzheimer's disease. Psychogeriatrics 2021; 21:659-667. [PMID: 33851473 DOI: 10.1111/psyg.12685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 01/20/2023]
Abstract
Phospholipid transfer protein (PLTP) is a complex glycosylated protein that mediates the transfer of phospholipids, unesterified cholesterol, diacylglycerides, specific apolipoproteins, and tocopherols between different classes of lipoproteins as well as between lipoproteins and cells. Many studies have associated PLTP with a variety of lipid metabolic diseases. However, recent studies have indicated that PLTP is highly expressed in the brain of vertebrate and may be related to many central nervous system diseases, such as Alzheimer's disease. Here, we review the data and report the role and mechanisms PLTP in Alzheimer's disease.
Collapse
Affiliation(s)
- Hao Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Mei-Hua Chen
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Wei Chen
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Ji-Guo Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Shu-Cun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong; Institute of Atherosclerosis, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| |
Collapse
|
7
|
Ma F, Darabi M, Lhomme M, Tubeuf E, Canicio A, Brerault J, Medadje N, Rached F, Lebreton S, Frisdal E, Brites F, Serrano C, Santos R, Gautier E, Huby T, El Khoury P, Carrié A, Abifadel M, Bruckert E, Guerin M, Couvert P, Giral P, Lesnik P, Le Goff W, Guillas I, Kontush A. Phospholipid transfer to high-density lipoprotein (HDL) upon triglyceride lipolysis is directly correlated with HDL-cholesterol levels and is not associated with cardiovascular risk. Atherosclerosis 2021; 324:1-8. [PMID: 33798922 DOI: 10.1016/j.atherosclerosis.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/05/2021] [Accepted: 03/04/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND AIMS While low concentrations of high-density lipoprotein-cholesterol (HDL-C) represent a well-established cardiovascular risk factor, extremely high HDL-C is paradoxically associated with elevated cardiovascular risk, resulting in the U-shape relationship with cardiovascular disease. Free cholesterol transfer to HDL upon lipolysis of triglyceride-rich lipoproteins (TGRL) was recently reported to underlie this relationship, linking HDL-C to triglyceride metabolism and atherosclerosis. In addition to free cholesterol, other surface components of TGRL, primarily phospholipids, are transferred to HDL during lipolysis. It remains indeterminate as to whether such transfer is linked to HDL-C and cardiovascular disease. METHODS AND RESULTS When TGRL was labelled with fluorescent phospholipid 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI), time- and dose-dependent transfer of DiI to HDL was observed upon incubations with lipoprotein lipase (LPL). The capacity of HDL to acquire DiI was decreased by -36% (p<0.001) in low HDL-C patients with acute myocardial infarction (n = 22) and by -95% (p<0.001) in low HDL-C subjects with Tangier disease (n = 7), unchanged in low HDL-C patients with Type 2 diabetes (n = 17) and in subjects with high HDL-C (n = 20), and elevated in subjects with extremely high HDL-C (+11%, p<0.05) relative to healthy normolipidemic controls. Across all the populations combined, HDL capacity to acquire DiI was directly correlated with HDL-C (r = 0.58, p<0.001). No relationship of HDL capacity to acquire DiI with both overall and cardiovascular mortality obtained from epidemiological studies for the mean HDL-C levels observed in the studied populations was obtained. CONCLUSIONS These data indicate that the capacity of HDL to acquire phospholipid from TGRL upon LPL-mediated lipolysis is proportional to HDL-C and does not reflect cardiovascular risk in subjects widely differing in HDL-C levels.
Collapse
Affiliation(s)
- Feng Ma
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France.
| | - Maryam Darabi
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Marie Lhomme
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, F-75013, France
| | - Emilie Tubeuf
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Aurélie Canicio
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Jean Brerault
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Narcisse Medadje
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Fabiana Rached
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France; Heart Institute-InCor, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Eric Frisdal
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Fernando Brites
- Laboratory of Lipids and Atherosclerosis, Department of Clinical Biochemistry, INFIBIOC, University of Buenos Aires, CONICET. Buenos Aires, Argentina
| | - Carlos Serrano
- Heart Institute-InCor, University of Sao Paulo, Sao Paulo, Brazil
| | - Raul Santos
- Heart Institute-InCor, University of Sao Paulo, Sao Paulo, Brazil
| | - Emmanuel Gautier
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Thierry Huby
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Petra El Khoury
- Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University, Beirut, Lebanon; INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
| | - Alain Carrié
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Marianne Abifadel
- Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University, Beirut, Lebanon; INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
| | - Eric Bruckert
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), Paris, F-75013, France; AP-HP, Groupe Hospitalier Pitié-Salpétrière, Paris, F-75013, France
| | - Maryse Guerin
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Philippe Couvert
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Philippe Giral
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), Paris, F-75013, France; AP-HP, Groupe Hospitalier Pitié-Salpétrière, Paris, F-75013, France
| | - Philippe Lesnik
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Wilfried Le Goff
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Isabelle Guillas
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Anatol Kontush
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| |
Collapse
|
8
|
Maglinger B, Frank JA, McLouth CJ, Trout AL, Roberts JM, Grupke S, Turchan-Cholewo J, Stowe AM, Fraser JF, Pennypacker KR. Proteomic changes in intracranial blood during human ischemic stroke. J Neurointerv Surg 2020; 13:395-399. [PMID: 32641418 PMCID: PMC7982920 DOI: 10.1136/neurintsurg-2020-016118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 01/29/2023]
Abstract
Background Since 2015, mechanical thrombectomy has been the standard treatment for emergent large vessel occlusion ischemic stroke. Objective To investigate, using the previously published Blood and Clot Thrombectomy Registry and Collaboration (BACTRAC) protocol (clinicaltrials.gov NCT03153683), how the protein expression of a patient’s intracranial blood during ischemic stroke compares with the protein expression of their systemic arterial blood in order to better understand and treat stroke. Methods Plasma samples from 25 subjects underwent proteomic analysis, where intracranial protein expression was compared with systemic protein levels. Data including sex, comorbidities, infarct volume, and infarct time were included for each subject. Results A majority of important proteins had a lower expression in intracranial blood than in systemic arterial blood. Proteins with the most significant changes in expression were: endopeptidase at −0.26 (p<0.0001), phospholipid transfer protein (PLTP) at −0.26 (p=0.0005), uromodulin (UMOD) at −0.14 (p=0.002), ficolin-2 (FCN2) at −0.46 (p=0.005), C-C motif chemokine 19 (CCL19) at −0.51 (p<0.0001), C-C motif chemokine 20 (CCL20) at −0.40 (p<0.0001), fibroblast growth factor 21 at −0.37 (p=0.0002), and C-C motif chemokine (CCL23) at −0.43 (p=0.0003). Conclusions Evaluation of proteomic changes in the intravascular space of a cerebral infarct in progress in human subjects suggested that changes in proteins such PLTP, fetuin-B (FETUB), and FCN2 may be involved in atherosclerotic changes, and chemokines such as CCL23 are known to play a role in the Th2 autoimmune response. These data provide a scientific springboard for identifying clinically relevant biomarkers for diagnosis/prognosis, and targets for much needed neuroprotective/neuroreparative pharmacotherapies.
Collapse
Affiliation(s)
- Benton Maglinger
- Department of Neurology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Jacqueline A Frank
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | | | - Amanda L Trout
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Jill Marie Roberts
- Department of Neurosurgery; Department of Neuroscience; Department of Neurology; Department of Radiology; Center for Advanced Translational Stroke Science, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Stephen Grupke
- Department of Neurosurgery; Department of Radiology; Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
| | - Jadwiga Turchan-Cholewo
- Department of Neurology; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Ann M Stowe
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Justin F Fraser
- Department of Neurosurgery; Department of Radiology; Department of Neuroscience; Department of Neurology; Department of Radiology; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Keith R Pennypacker
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
9
|
Chen X, Wang S, Huang Y, Zhao X, Jia X, Meng G, Zheng Q, Zhang M, Wu Y, Wang L. Obesity Reshapes Visceral Fat-Derived MHC I Associated-Immunopeptidomes and Generates Antigenic Peptides to Drive CD8 + T Cell Responses. iScience 2020; 23:100977. [PMID: 32217358 PMCID: PMC7109454 DOI: 10.1016/j.isci.2020.100977] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/21/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Adaptive CD8+ T cells were observed to contribute to the initiation and progression of obesity-induced visceral adipose tissue (VAT) chronic inflammation that is critically linked to metabolic disorders. Numerous peptides presented by the major histocompatibility complex (MHC) class I molecules at the cell surface are collectively termed as MHC I-associated immunopeptidome (MIP) for the interaction with CD8+ T cells. We conducted the in-depth mapping of MIP of VAT from lean and obese mice using large-scale high-resolution mass spectrometry and observed that obesity significantly alters the landscape of VAT MIPs. Additionally, the obese VAT-exclusive MIP source proteome reflected a distinct obesity-associated signature. A peptide derived from lactate dehydrogenase A (LDHA) or B chain, named LDHA237-244, was identified as an obese VAT-exclusive immunogenic peptide that was capable of eliciting pro-inflammatory CD8+ T cells responses. Our findings suggest that certain immunogenic peptides generated by obesity may trigger CD8+ T cell-mediated VAT inflammation. Obesity reshapes the landscape of VAT-derived MIP The obese VAT-exclusive MIP reflects an obesity-associated signature An obese VAT-exclusive peptide LDHA237-244 can stimulate CD8+ T cell responses LDHA237-244-reactive CD8+ T cells were present in obese mice but not lean mice
Collapse
Affiliation(s)
- Xiaoling Chen
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shufeng Wang
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yi Huang
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xia Zhao
- Bioinformatics Center, Department of Microbiology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xu Jia
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Physiology, North Sichuan Medical College, Nanchong 637007, China
| | - Gang Meng
- Department of Pathology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qian Zheng
- Department of Physiology, North Sichuan Medical College, Nanchong 637007, China
| | - Mengjun Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuzhang Wu
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Li Wang
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
10
|
Cai Z, Yu C, Fu D, Pan Y, Huang J, Rong Y, Deng L, Chen J, Chen M. Differential metabolic and hepatic transcriptome responses of two miniature pig breeds to high dietary cholesterol. Life Sci 2020; 250:117514. [PMID: 32145306 DOI: 10.1016/j.lfs.2020.117514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022]
Abstract
AIMS Pigs are increasingly used as human metabolic disease models; however, there is insufficient research on breed-related genetic background differences. This study aimed to investigate the differential metabolic responses to high-fat and high-cholesterol (HFC) diet-induced non-alcoholic fatty liver disease (NAFLD) of two miniature pig breeds and explore the molecular mechanisms involved. MAIN METHODS Male Wuzhishan (WZSP) and Tibetan pigs (TP) were randomly fed either a standard or an HFC diet for 24 weeks. Weight, serum lipids, bile acid, insulin resistance, liver function, liver histology, and hepatic lipid deposition were determined. RNA-Seq was used to detect the hepatic gene expression profiles. Western blot, immunohistochemistry, and qRT-PCR were used to detect the lipid and glucose metabolism-related gene expressions. KEY FINDINGS The HFC diet caused obesity, hypertension, severe hypercholesterolemia, liver injury, increased hepatocellular steatosis and inflammation, and significantly increased serum insulin levels in both pig breeds. This diet led to higher serum and hepatic cholesterol level concentrations in WZSP and elevated fasting glucose levels in TP. Transcriptome analysis revealed that the genes controlling hepatic cholesterol metabolism and the inflammatory response were consistently regulated; lipid metabolism and insulin signaling related genes were uniquely regulated by the HFC diet in the WZSP and TP, respectively. SIGNIFICANCE Our study demonstrated that the genetic background affects profoundly pigs' metabolic and hepatic responses to an HFC diet. These results deepened our understanding of the molecular mechanisms of HFC diet-induced NAFLD and provided a foundation for selecting the appropriate pig breeds for metabolic studies in the future.
Collapse
Affiliation(s)
- Zhaowei Cai
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chen Yu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Danting Fu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yongming Pan
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Junjie Huang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yili Rong
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liqun Deng
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiaojiao Chen
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Minli Chen
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
11
|
Antikainen AAV, Sandholm N, Trégouët DA, Charmet R, McKnight AJ, Ahluwalia TS, Syreeni A, Valo E, Forsblom C, Gordin D, Harjutsalo V, Hadjadj S, Maxwell AP, Rossing P, Groop PH. Genome-wide association study on coronary artery disease in type 1 diabetes suggests beta-defensin 127 as a risk locus. Cardiovasc Res 2020; 117:600-612. [PMID: 32077919 DOI: 10.1093/cvr/cvaa045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/20/2019] [Accepted: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
AIMS Diabetes is a known risk factor for coronary artery disease (CAD). There is accumulating evidence that CAD pathogenesis differs for individuals with type 1 diabetes (T1D). However, the genetic background has not been extensively studied. We aimed to discover genetic loci increasing CAD susceptibility, especially in T1D, to examine the function of these discoveries and to study the role of the known risk loci in T1D. METHODS AND RESULTS We performed the largest genome-wide association study to date for CAD in T1D, comprising 4869 individuals with T1D (cases/controls: 941/3928). Two loci reached genome-wide significance, rs1970112 in CDKN2B-AS1 [odds ratio (OR) = 1.32, P = 1.50 × 10-8], and rs6055069 on DEFB127 promoter (OR = 4.17, P = 2.35 × 10-9), with consistent results in survival analysis. The CDKN2B-AS1 variant replicated (P = 0.04) when adjusted for diabetic kidney disease in three additional T1D cohorts (cases/controls: 434/3123). Furthermore, we explored the function of the lead discoveries with a cardio-phenome-wide analysis. Among the eight suggestive loci (P < 1 × 10-6), rs70962766 near B3GNT2 associated with central blood pressure, rs1344228 near CNTNAP5 with intima media thickness, and rs2112481 on GRAMD2B promoter with serum leucocyte concentration. Finally, we calculated genetic risk scores for individuals with T1D with the known susceptibility loci. General population risk variants were modestly but significantly associated with CAD also in T1D (P = 4.21 × 10-7). CONCLUSION While general population CAD risk loci had limited effect on the risk in T1D, for the first time, variants at the CDKN2B-AS1 locus were robustly associated with CAD in individuals with T1D. The novel finding on β-defensin DEFB127 promoter provides a link between diabetes, infection susceptibility, and CAD, although pending on future confirmation.
Collapse
Affiliation(s)
- Anni A V Antikainen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland
| | - David-Alexandre Trégouët
- Sorbonne Université, UPMC Univ Paris 06, INSERM UMR_S 1166, Paris, France.,ICAN Institute for Cardiometabolism and Nutrition, Paris, France.,INSERM UMR_S 1219, Bordeaux Population Health Research Center, Bordeaux University, Bordeaux, France
| | - Romain Charmet
- Sorbonne Université, UPMC Univ Paris 06, INSERM UMR_S 1166, Paris, France.,ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Amy Jayne McKnight
- Centre for Public Health, Queens University of Belfast, Northern Ireland BT7 1NN, UK
| | | | - Anna Syreeni
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland
| | - Erkka Valo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland
| | - Daniel Gordin
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland.,Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland.,The Chronic Disease Prevention Unit, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Samy Hadjadj
- Department of Endocrinology and Diabetology, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,INSERM CIC 1402, Poitiers, France.,L'institut du thorax, INSERM, CNRS, UNIV, Nantes CHU Nantes, Nantes, France
| | - Alexander P Maxwell
- Centre for Public Health, Queens University of Belfast, Northern Ireland BT7 1NN, UK
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, DK 2820 Gentofte, Denmark.,University of Copenhagen, Copenhagen, Denmark
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, FI-00290 Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, FI-00290 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00290 Helsinki, Finland.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Dupas S, Neiers F, Granon E, Rougeux E, Dupont S, Beney L, Bousquet F, Shaik HA, Briand L, Wojtasek H, Charles JP. Collisional mechanism of ligand release by Bombyxmori JHBP, a member of the TULIP / Takeout family of lipid transporters. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 117:103293. [PMID: 31809784 DOI: 10.1016/j.ibmb.2019.103293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 06/10/2023]
Abstract
Juvenile hormones (JHs) regulate important processes in insects, such as postembryonic development and reproduction. In the hemolymph of Lepidoptera, these lipophilic sesquiterpenic hormones are transported from their site of synthesis to target tissues by high affinity carriers, the juvenile hormone binding proteins (JHBPs). Lepidopteran JHBPs belong to a recently uncovered, yet very ancient family of proteins sharing a common lipid fold (TULIP domain) and involved in shuttling various lipid ligands. One important, but poorly understood aspect of JHs action, is the mechanism of hormone transfer to or through the plasma membranes of target cells. Since many membrane-active peptides and proteins, such as the pore-forming bacterial toxins, are activated by low pH or interaction with phospholipid membranes, we have examined the effect of these factors on JH binding by JHBPs. The affinity of Bombyx mori and Manduca sexta JHBPs for JH III was determined by the DCC assay, equilibrium dialysis, and isothermal titration calorimetry, and found to be greatly reduced at low pH, in agreement with previous observations. Loss of binding was accompanied by changes in fluorescence and near-UV CD spectra, indicating significant changes in protein structure in the environment of aromatic residues. The apparent dissociation rate constant (koff) of the JHBP-JH III complex was greater at acidic pH, suggesting that low pH favors ligand release by opening of the binding pocket. The affinity of recombinant B. mori JHBP (rBmJHBP) was also decreased in the presence of anionic phospholipid vesicles. Measurements of steady-state fluorescence anisotropy with the lipophilic probe TMA-DPH demonstrated that rBmJHBP specifically interacts with anionic membranes. These results suggest the existence of a collisional mechanism for ligand release that may be important for delivery of JHs to the target cells, and could be relevant to the function of related members of this emerging family of lipid-transport proteins.
Collapse
Affiliation(s)
- Stéphane Dupas
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Fabrice Neiers
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Emma Granon
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Erwan Rougeux
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Sébastien Dupont
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - Laurent Beney
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - François Bousquet
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Haq Abdul Shaik
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Loic Briand
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Hubert Wojtasek
- Institute of Chemistry, Opole University, Ul. Oleska 48, 45-052, Opole, Poland.
| | - Jean-Philippe Charles
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France.
| |
Collapse
|
13
|
Alterations of lipid metabolism, blood pressure and fatty liver in spontaneously hypertensive rats transgenic for human cholesteryl ester transfer protein. Hypertens Res 2020; 43:655-666. [DOI: 10.1038/s41440-020-0401-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/08/2019] [Accepted: 01/07/2020] [Indexed: 02/08/2023]
|
14
|
van Dijk PR, Abdulle AE, Bulthuis ML, Perton FG, Connelly MA, van Goor H, Dullaart RP. The Systemic Redox Status Is Maintained in Non-Smoking Type 2 Diabetic Subjects Without Cardiovascular Disease: Association with Elevated Triglycerides and Large VLDL. J Clin Med 2019; 9:jcm9010049. [PMID: 31878321 PMCID: PMC7019670 DOI: 10.3390/jcm9010049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/10/2019] [Accepted: 12/22/2019] [Indexed: 12/11/2022] Open
Abstract
Decreased circulating levels of free thiols (R-SH, sulfhydryl groups) reflect enhanced oxidative stress, which plays an important role in the pathogenesis of cardiometabolic diseases. Since hyperglycemia causes oxidative stress, we questioned whether plasma free thiols are altered in patients with type 2 diabetes mellitus (T2DM) without cardiovascular disease or renal function impairment. We also determined their relationship with elevated triglycerides and very low density lipoproteins (VLDL), a central feature of diabetic dyslipidemia. Fasting plasma free thiols (colorimetric method), lipoproteins, VLDL (nuclear magnetic resonance spectrometry), free fatty acids (FFA), phospholipid transfer protein (PLTP) activity and adiponectin were measured in 79 adult non-smoking T2DM subjects (HbA1c 51 ± 8 mmol/mol, no use of insulin or lipid lowering drugs), and in 89 non-smoking subjects without T2DM. Plasma free thiols were univariately correlated with glucose (r = 0.196, p < 0.05), but were not decreased in T2DM subjects versus non-diabetic subjects (p = 0.31). Free thiols were higher in subjects with (663 ± 84 µmol/L) versus subjects without elevated triglycerides (619 ± 91 µmol/L; p = 0.002). Age- and sex-adjusted multivariable linear regression analysis demonstrated that plasma triglycerides were positively and independently associated with free thiols (β = 0.215, p = 0.004), FFA (β = 0.168, p = 0.029) and PLTP activity (β = 0.228, p = 0.002), inversely with adiponectin (β = −0.308, p < 0.001) but not with glucose (β = 0.052, p = 0.51). Notably, the positive association of free thiols with (elevated) triglycerides appeared to be particularly evident in men. Additionally, large VLDL were independently associated with free thiols (β = 0.188, p = 0.029). In conclusion, circulating free thiols are not decreased in this cohort of non-smoking and generally well-controlled T2DM subjects. Paradoxically, higher triglycerides and more large VLDL particles are likely associated with higher plasma levels of thiols, reflecting lower systemic oxidative stress.
Collapse
Affiliation(s)
- Peter R. van Dijk
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
- Correspondence:
| | - Amaal Eman Abdulle
- Department of Internal Medicine, division vascular medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Marian L.C. Bulthuis
- Department of Pathology and Medical, Biology, Section Pathology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands (H.v.G.)
| | - Frank G. Perton
- Laboratory Center, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Margery A. Connelly
- Laboratory Corporation of America® Holdings (LabCorp), Morrisville, NC 27560, USA;
| | - Harry van Goor
- Department of Pathology and Medical, Biology, Section Pathology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands (H.v.G.)
| | - Robin P.F. Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| |
Collapse
|
15
|
Song G, Zong C, Shao M, Yu Y, Liu Q, Wang H, Qiu T, Jiao P, Guo Z, Lee P, Luo Y, Jiang XC, Qin S. Phospholipid transfer protein (PLTP) deficiency attenuates high fat diet induced obesity and insulin resistance. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1305-1313. [PMID: 31220615 DOI: 10.1016/j.bbalip.2019.06.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/07/2019] [Accepted: 06/14/2019] [Indexed: 01/09/2023]
Abstract
Increased phospholipid transfer protein (PLTP) activity has been found to be associated with obesity, and metabolic syndrome in humans. However, whether or not PLTP has a direct effect on insulin sensitivity and obesity is largely unknown. Here we analyzed the effect by using PLTP knockout (PLTP-/-) mouse model. Although, PLTP-/- mice have normal body-weight-gain under chow diet, these mice were protected from high-fat-diet-induced obesity and insulin resistance, compared with wild type mice. In order to understand the mechanism, we evaluated insulin receptor and Akt activation and found that PLTP deficiency significantly enhanced phosphorylated insulin receptor and Akt levels in high-fat-diet fed mouse livers, adipose tissues, and muscles after insulin stimulation, while total Akt and insulin receptor levels were unchanged. Moreover, we found that the PLTP deficiency induced significantly more GLUT4 protein in the plasma membranes of adipocytes and muscle cells after insulin stimulation. Finally, we found that PLTP-deficient hepatocytes had less sphingomyelins and free cholesterols in the lipid rafts and plasma membranes than that of controls and this may provide a molecular basis for PLTP deficiency-mediated increase in insulin sensitivity. We have concluded that PLTP deficiency leads to an improvement in tissue and whole-body insulin sensitivity through modulating lipid levels in the plasma membrane, especially in the lipid rafts.
Collapse
Affiliation(s)
- Guohua Song
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China.
| | - Chuanlong Zong
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Mingzhu Shao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Yang Yu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Qian Liu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Hui Wang
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Tingting Qiu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Zheng Guo
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Phoebe Lee
- Downstate Medical Center State University of New York, NY, USA
| | - Yi Luo
- Downstate Medical Center State University of New York, NY, USA
| | | | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China.
| |
Collapse
|
16
|
Jing R, Liu Y, Guo P, Ni T, Gao X, Mei R, He X, Zhang J. Evaluation of Common Variants in Matrix Metalloproteinase-9 Gene with Lumbar Disc Herniation in Han Chinese Population. Genet Test Mol Biomarkers 2018; 22:622-629. [PMID: 30289281 DOI: 10.1089/gtmb.2018.0080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Lumbar disc herniation (LDH) is a common and frequent orthopedic disease with strong genetic determinants. The disruption of the intervertebral disc extracellular matrix has been found to play a key role in the development of LDH, suggesting that abnormal matrix metalloproteinases (MMPs) may promote the degradation of the disc matrix. MMP-9, an important member of the MMP family, is a good candidate for the LDH susceptibility gene. The present study aimed to investigate the association of common variants in the MMP-9 gene with the risk, severity, and clinical characteristic variables of LDH. MATERIALS AND METHODS Fourteen tag single nucleotide polymorphisms (SNPs) entirely covering the region of the MMP-9 gene were analyzed in a sample of 845 patients and 1751 healthy controls. RESULTS The SNP rs17576 was found to be significantly associated with susceptibility to LDH (OR = 0.77, p = 0.0002), which was also confirmed by haplotype-based analyses (rs79845319-rs17576-rs45437897, global p < 0.001). Our results indicated that the A allele of rs17576 reduced the risk of LDH by ∼23% on average. Furthermore, the G allele of rs17576 was found to correlate with more severe grades of disc degeneration. CONCLUSION Our results provide additional evidence supporting an important role of the MMP-9 gene in the pathogenesis of LDH.
Collapse
Affiliation(s)
- Rong Jing
- 1 Department of Orthopedics, The Second Affiliated Hospital, Xi'an Jiaotong University , Xi'an, China
| | - Yunlei Liu
- 2 Department of Traditional Chinese Medicine, Affiliated Hospital of Yan'an University , Yan' an, China
| | - Peng Guo
- 3 Department of Joint Surgery, Yan'an People's Hospital , Yan'an, China
| | - Tong Ni
- 4 Key Laboratory of National Ministry of Health for Forensic Sciences, School of Medicine and Forensics, Xi'an Jiaotong University , Xi'an, China
| | - Xiang Gao
- 5 Department of Rehabilitation Medicine, Affiliated Hospital of Yan'an University , Yan' an, China
| | - Rong Mei
- 5 Department of Rehabilitation Medicine, Affiliated Hospital of Yan'an University , Yan' an, China
| | - Xijing He
- 1 Department of Orthopedics, The Second Affiliated Hospital, Xi'an Jiaotong University , Xi'an, China
| | - Jianlin Zhang
- 3 Department of Joint Surgery, Yan'an People's Hospital , Yan'an, China
| |
Collapse
|
17
|
Nass KJ, van den Berg EH, Gruppen EG, Dullaart RPF. Plasma lecithin:cholesterol acyltransferase and phospholipid transfer protein activity independently associate with nonalcoholic fatty liver disease. Eur J Clin Invest 2018; 48:e12988. [PMID: 29947103 DOI: 10.1111/eci.12988] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent condition which contributes to atherogenic apolipoprotein B dyslipoproteinemias. Lecithin:cholesterol acyltransferase (LCAT) and phospholipid transfer protein (PLTP) are both synthesized by the liver and are important in lipid metabolism. Here, we interrogated the impact of NAFLD on plasma LCAT and PLTP activities. METHODS Plasma LCAT activity (exogenous substrate assay) and PLTP activity (phospholipid vesicles-HDL assay) were determined in 348 subjects (279 men; 81 subjects with type 2 diabetes (T2DM); 123 with metabolic syndrome (MetS)). A Fatty Liver Index (FLI) ≥60 was used as a proxy of NAFLD. Insulin resistance was determined by homoeostasis model assessment (HOMA-IR). RESULTS A total of 147 participants had an FLI ≥60 coinciding with T2DM and MetS (P < 0.001 for each). Plasma LCAT activity and PLTP activity were on average 12% and 5% higher, respectively, in subjects with an FLI ≥ 60 (P < 0.001 for each). In age- and sex-adjusted partial linear regression analysis, LCAT activity and PLTP activity were positively related to various obesity measures and HOMA-IR (P < 0.001 for each). In multivariable linear regression analyses adjusted for age and sex, LCAT activity was associated with an FLI ≥ 60 independent of T2DM and MetS, the waist/hip ratio, or HOMA-IR (β = 0.307 to 0.366, P < 0001 for all models). PLTP activity was also associated with an FLI ≥ 60 independent of these variables (β = 0.151 to 0223, P = 0.013 to 0.001). CONCLUSION NAFLD, as inferred from an FLI≥60, confers higher plasma LCAT and to a lesser extent PLTP activity, even when taking account of T2DM, MetS, central obesity and insulin resistance.
Collapse
Affiliation(s)
- Karlijn J Nass
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eline H van den Berg
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eke G Gruppen
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
18
|
Plasma phospholipid transfer protein (PLTP) as an emerging determinant of the adaptive immune response. Cell Mol Immunol 2018; 15:1077-1079. [PMID: 29735978 DOI: 10.1038/s41423-018-0036-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 04/12/2018] [Indexed: 11/08/2022] Open
|
19
|
Deletion of plasma Phospholipid Transfer Protein (PLTP) increases microglial phagocytosis and reduces cerebral amyloid-β deposition in the J20 mouse model of Alzheimer's disease. Oncotarget 2018; 9:19688-19703. [PMID: 29731975 PMCID: PMC5929418 DOI: 10.18632/oncotarget.24802] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/27/2018] [Indexed: 01/22/2023] Open
Abstract
Plasma phospholipid transfer protein (PLTP) binds and transfers a number of amphipathic compounds, including phospholipids, cholesterol, diacylglycerides, tocopherols and lipopolysaccharides. PLTP functions are relevant for many pathophysiological alterations involved in neurodegenerative disorders (especially lipid metabolism, redox status, and immune reactions), and a significant increase in brain PLTP levels was observed in patients with Alzheimer's disease (AD) compared to controls. To date, it has not been reported whether PLTP can modulate the formation of amyloid plaques, i.e. one of the major histopathological hallmarks of AD. We thus assessed the role of PLTP in the AD context by breeding PLTP-deficient mice with an established model of AD, the J20 mice. A phenotypic characterization of the amyloid pathology was conducted in J20 mice expressing or not PLTP. We showed that PLTP deletion is associated with a significant reduction of cerebral Aβ deposits and astrogliosis, which can be explained at least in part by a rise of Aβ clearance through an increase in the microglial phagocytic activity and the expression of the Aβ-degrading enzyme neprilysin. PLTP arises as a negative determinant of plaque clearance and over the lifespan, elevated PLTP activity could lead to a higher Aβ load in the brain.
Collapse
|
20
|
Gruppen EG, Kersten S, Dullaart RPF. Plasma angiopoietin-like 4 is related to phospholipid transfer protein activity in diabetic and non-diabetic subjects: role of enhanced low grade inflammation. Lipids Health Dis 2018; 17:60. [PMID: 29587751 PMCID: PMC5870514 DOI: 10.1186/s12944-018-0717-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/21/2018] [Indexed: 01/22/2023] Open
Abstract
Background Angiopoietin-like 4 (ANGPTL4) inhibits lipoprotein lipase, whereas phospholipid transfer protein (PLTP) enhances hepatic triglyceride secretion. Both factors may be upregulated by inflammatory pathways. Since the extent to which these circulating factors are interrelated is unknown, we determined the relationship between plasma ANGPTL4 and PLTP activity, and assessed whether such a relationship could be explained by high sensitivity C-reactive protein (hsCRP) levels as a marker of low-grade chronic inflammation. Methods Fasting plasma ANGPTL4, PLTP activity (liposome-vesicle high density lipoprotein system) and hsCRP were measured in 41 type 2 diabetic (T2DM) subjects and 36 non-diabetic subjects. Results Plasma ANGPTL4 and PLTP activity were increased in T2DM (p < 0.001 for each), coinciding with elevated hsCRP, triglycerides and non-esterified fatty acids (NEFA) (p = 0.031 to 0.001). In univariate analysis, ANGTLP4 was correlated with PLTP activity (Rs = 0.309, p = 0.006), whereas both factors were related to hsCRP and NEFA levels (Rs = 0.304 to 0.411, p < 0.01 to < 0.001). In multivariable linear regression analysis adjusting for age, sex, glucose, total cholesterol, triglycerides and NEFA, ANGPTL4 and PLTP activity each remained positively associated with hsCRP (β = 0.315, p = 0.003 and β = 0.299, p = 0.034, respectively). Plasma ANGPTL4 remained positively associated with PLTP activity when taking account of age, sex, glucose, total cholesterol, triglycerides and NEFA (β = 0.315, p = 0.003). Notably, this association disappeared after further adjustment for hsCRP (β = 0.131, p = 0.25). Conclusions In conclusion, plasma ANGPTL4 and PLTP activity are interrelated, which may at least in part be explained by low-grade chronic inflammation. A pro-inflammatory state could affect triglyceride metabolism via concerted effects on ANGPTL4 and PLTP.
Collapse
Affiliation(s)
- Eke G Gruppen
- Department of Endocrinology, University of Groningen and University Medical Center, P.O. Box 301, 9700 RB, Groningen, The Netherlands
| | - Sander Kersten
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen and University Medical Center, P.O. Box 301, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
21
|
Perrier V, Imberdis T, Lafon PA, Cefis M, Wang Y, Huetter E, Arnaud JD, Alvarez-Martinez T, Le Guern N, Maquart G, Lagrost L, Desrumaux C. Plasma cholesterol level determines in vivo prion propagation. J Lipid Res 2017; 58:1950-1961. [PMID: 28765208 PMCID: PMC5625119 DOI: 10.1194/jlr.m073718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 07/28/2017] [Indexed: 12/27/2022] Open
Abstract
Transmissible spongiform encephalopathies are fatal neurodegenerative diseases with an urgent need for therapeutic and prophylactic strategies. At the time when the blood-mediated transmission of prions was demonstrated, in vitro studies indicated a high binding affinity of the scrapie prion protein (PrPSc) with apoB-containing lipoproteins, i.e., the main carriers of cholesterol in human blood. The aim of the present study was to explore the relationship between circulating cholesterol-containing lipoproteins and the pathogenicity of prions in vivo. We showed that, in mice with a genetically engineered deficiency for the plasma lipid transporter, phospholipid transfer protein (PLTP), abnormally low circulating cholesterol concentrations were associated with a significant prolongation of survival time after intraperitoneal inoculation of the 22L prion strain. Moreover, when circulating cholesterol levels rose after feeding PLTP-deficient mice a lipid-enriched diet, a significant reduction in survival time of mice together with a marked increase in the accumulation rate of PrPSc deposits in their brain were observed. Our results suggest that the circulating cholesterol level is a determinant of prion propagation in vivo and that cholesterol-lowering strategies might be a successful therapeutic approach for patients suffering from prion diseases.
Collapse
Affiliation(s)
- Véronique Perrier
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Thibaud Imberdis
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Pierre-André Lafon
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Marina Cefis
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Yunyun Wang
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France.,Cellular Signaling Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Elisabeth Huetter
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Jacques-Damien Arnaud
- Etablissement Confiné d'Expérimentation A3/L3, CECEMA, US009 Biocampus, UMS 3426, Université Montpellier, Montpellier, F-34095 France
| | - Teresa Alvarez-Martinez
- Etablissement Confiné d'Expérimentation A3/L3, CECEMA, US009 Biocampus, UMS 3426, Université Montpellier, Montpellier, F-34095 France
| | - Naig Le Guern
- INSERM, LNC UMR866, F-21000 Dijon, France and LNC UMR866, Université Bourgogne Franche-Comté, F-21000 Dijon, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France
| | - Guillaume Maquart
- INSERM, LNC UMR866, F-21000 Dijon, France and LNC UMR866, Université Bourgogne Franche-Comté, F-21000 Dijon, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France
| | - Laurent Lagrost
- INSERM, LNC UMR866, F-21000 Dijon, France and LNC UMR866, Université Bourgogne Franche-Comté, F-21000 Dijon, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France.,University Hospital of Dijon, F-21000 Dijon, France
| | - Catherine Desrumaux
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France .,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France
| |
Collapse
|
22
|
Skoczyńska A, Wojakowska A, Turczyn B, Zatońska K, Wołyniec M, Rogala N, Szuba A, Bednarek-Tupikowska G. Serum Lipid Transfer Proteins in Hypothyreotic Patients Are Inversely Correlated with Thyroid-Stimulating Hormone (TSH) Levels. Med Sci Monit 2016; 22:4661-4669. [PMID: 27899788 PMCID: PMC5144931 DOI: 10.12659/msm.898134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Plasma cholesteryl ester transfer protein (CETP) activity is often decreased in patients with hypothyroidism, whereas less is known about the phospholipid transfer protein (PLTP). We aimed to evaluate simultaneously serum CETP and PLTP activity in patients diagnosed with hypothyroidism. Material/Methods The selection criteria for control group members (without thyroid dysfunction) in this case to case study were levels of total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol (HDL-C), and triglycerides similar to those in study group patients (101 patients diagnosed with hypothyroidism). Serum CETP and PLTP activities were measured by homogenous fluorometric assays using synthetic donor particle substrates. Results Serum CETP and PLTP activities in hypothyreotic patients were lower (p<0.001) compared with those in healthy subjects. This lowering was associated with significant changes in HDL-C subclasses: decrease in HDL2- and increase in HDL3 cholesterol levels. Multiple linear regression analyses adjusted for age, sex, body mass index, smoking habits, and alcohol drinking showed a strong association between hypothyroidism and activity of lipid transfer proteins. A linear inverse relationship between thyroid-stimulating hormone (TSH) and CETP (r=−0.21; p<0.01) and between TSH and PLTP (r=−0.24; p<0.001) was shown. There also was a positive correlation (p<0.001) between CETP and HDL2 cholesterol (r=0.27) and between PLTP and HDL2 cholesterol (r=0.37). A negative correlation between CETP and HDL3 cholesterol (r=−0.22: p<0.01) and between PLTP and HDL3 cholesterol (r=−0.24; p<0.001) has been demonstrated as well. Conclusions The decreased HDL2 and increased HDL3 cholesterol levels in subjects with hypothyroidism are consequences of decreased activity of lipid transfer proteins. These changes are early symptoms of lipid disturbances in hypothyroidism.
Collapse
Affiliation(s)
- Anna Skoczyńska
- Department of Internal and Occupational Medicine and Hypertension, Wrocław Medical University, Wrocław, Poland
| | - Anna Wojakowska
- Department of Internal and Occupational Diseases, Wrocław Medical University, Wrocław, Poland
| | - Barbara Turczyn
- Department of Internal and Occupational Diseases, Wrocław Medical University, Wrocław, Poland
| | - Katarzyna Zatońska
- Department of Social Medicine, Wrocław Medical University, Wrocław, Poland
| | - Maria Wołyniec
- Department of Social Medicine, Wrocław Medical University, Wrocław, Poland
| | - Natalia Rogala
- Department of Endocrinology, Diabetology and Isotope Treatment, Wrocław Medical University, Wrocław, Poland
| | - Andrzej Szuba
- Department of Angiology, Wrocław Medical University, Wrocław, Poland
| | | |
Collapse
|
23
|
Skoczyńska A, Wojakowska A, Turczyn B, Zatońska K, Wołyniec M, Szuba A. Serum CETP and PLTP activity in middle-aged men living in urban or rural area of the Lower Silesia region. PURE Poland sub-study. Arch Med Sci 2016; 12:704-14. [PMID: 27478449 PMCID: PMC4947617 DOI: 10.5114/aoms.2016.60950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/09/2015] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The dependence of lipid transfer proteins on significant pro-atherogenic factors is unclear. The aim of the study was to evaluate serum cholesteryl ester transfer protein (CETP) and phospholipid transfer protein (PLTP) activity in relation to lipid disturbances in men living in an urban or rural area. MATERIAL AND METHODS A group of 427 men, volunteers for the Prospective Urban Rural Epidemiology (PURE) sub-study - 263 urban inhabitants (aged 51.9 ±6.0) and 164 residents of villages (aged 51.1 ±5.9) - were examined. In the multivariable linear regression model, the following factors were included as potential confounders: age, body mass index (BMI), smoking, alcohol consumption, hs-C-reactive protein reaction (hs-CRP) and co-existence of chronic diseases. RESULTS In multiple linear regression models, site of residence (urban or rural area) was the most important independent and consistent predictor of CETP and PLTP activity; β coefficients (95% CI) for CETP (0.18) and PLTP (-0.29) were significant at levels of p < 0.001. Three-way analysis of variance showed no effect of smoking or moderate alcohol consumption on lipid transfer proteins; however, CETP activity showed an interaction effect between these risk factors. In the group of all men, CETP activity was significantly and positively correlated with total cholesterol (r = 0.24), low-density lipoprotein cholesterol (r = 0.18), and non-high density lipoprotein cholesterol (r = 0.21), whereas PLTP activity was correlated with BMI (r = 0.12). Body mass index in rural men was higher than in the urban male population. CONCLUSIONS Increased PLTP activity, recognized as a pro-atherogenic factor, and decreased CETP activity, known as a protective factor, both observed in men living in rural areas, are probably conditioned by nutritional and/or genetic factors.
Collapse
Affiliation(s)
- Anna Skoczyńska
- Department of Internal and Occupational Diseases and Hypertension, Wroclaw Medical University, Wroclaw, Poland
| | - Anna Wojakowska
- Department of Internal and Occupational Diseases and Hypertension, Wroclaw Medical University, Wroclaw, Poland
| | - Barbara Turczyn
- Department of Internal and Occupational Diseases and Hypertension, Wroclaw Medical University, Wroclaw, Poland
| | - Katarzyna Zatońska
- Department of Internal and Occupational Diseases and Hypertension, Wroclaw Medical University, Wroclaw, Poland
| | - Maria Wołyniec
- Department of Internal and Occupational Diseases and Hypertension, Wroclaw Medical University, Wroclaw, Poland
| | - Andrzej Szuba
- Department of Internal and Occupational Diseases and Hypertension, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
24
|
Thomas Jennings R, Odkhuu E, Nakashima A, Morita N, Kobayashi T, Yamai I, Tanaka M, Suganami T, Haga S, Ozaki M, Watanabe Y, Nagai Y, Takatsu K, Kikuchi-Ueda T, Ichimonji I, Ogawa Y, Takagi H, Yamazaki T, Miyake K, Akashi-Takamura S. Inflammatory responses increase secretion of MD-1 protein. Int Immunol 2016; 28:503-512. [PMID: 27352793 DOI: 10.1093/intimm/dxw031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 06/21/2016] [Indexed: 12/16/2022] Open
Abstract
Radioprotective 105 (RP105) is a type I transmembrane protein, which associates with a glycoprotein, MD-1. Monoclonal antibody (mAb)-mediated ligation of RP105/MD-1 robustly activates B cells. RP105/MD-1 is structurally similar to Toll-like receptor 4 (TLR4)/MD-2. B-cell responses to TLR2 and TLR4/MD-2 ligands are impaired in the absence of RP105 or MD-1. In addition to RP105/MD-1, MD-1 alone is secreted. The structure of MD-1 shows that MD-1 has a hydrophobic cavity that directly binds to phospholipids. Little is known, however, about a ligand for MD-1 and the role of MD-1 in vivo To study the role of RP105/MD-1 and MD-1 alone, specific mAbs against MD-1 are needed. Here, we report the establishment and characterization of two anti-MD-1 mAbs (JR2G9, JR7G1). JR2G9 detects soluble MD-1, whereas JR7G1 binds both soluble MD-1 and the cell surface RP105/MD-1 complex. With these mAbs, soluble MD-1 was detected in the serum and urine. The MD-1 concentration was altered by infection, diet and reperfusion injury. Serum MD-1 was rapidly elevated by TLR ligand injection in mice. The quantitative PCR and supernatant-precipitated data indicate that macrophages are one of the sources of serum soluble MD-1. These results suggest that soluble MD-1 is a valuable biomarker for inflammatory diseases.
Collapse
Affiliation(s)
- Richard Thomas Jennings
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Erdenezaya Odkhuu
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Akina Nakashima
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Naoko Morita
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Toshihiko Kobayashi
- Department of Molecular Immunology and Inflammation, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjyuku-ku, Tokyo 162-8655, Japan
| | - Ikuko Yamai
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Sanae Haga
- Laboratory of Molecular and Functional Bio-imaging, Faculty of Health Sciences, Hokkaido University, Kita-ku, Sapporo, Hokkaido 060-0812, Japan
| | - Michitaka Ozaki
- Laboratory of Molecular and Functional Bio-imaging, Faculty of Health Sciences, Hokkaido University, Kita-ku, Sapporo, Hokkaido 060-0812, Japan
| | - Yasuharu Watanabe
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630, Toyama 930-0194, Japan
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630, Toyama 930-0194, Japan JST, PRESTO, 4-1-8, Kawaguchi, Saitama 332-0012, Japan
| | - Kiyoshi Takatsu
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630, Toyama 930-0194, Japan Toyama Prefectural Institute for Pharmaceutical Research, 17-1, Irimizu, Toyama 939-0363, Japan
| | - Takane Kikuchi-Ueda
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan
| | - Isao Ichimonji
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Bunkyo-ku, Tokyo 113-8510, Japan AMED, CREST, 1-7-1, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Hidekazu Takagi
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Tatsuya Yamazaki
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Sachiko Akashi-Takamura
- Department of Microbiology and Immunology, School of Medicine, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
25
|
Ponziani FR, Pecere S, Gasbarrini A, Ojetti V. Physiology and pathophysiology of liver lipid metabolism. Expert Rev Gastroenterol Hepatol 2016; 9:1055-67. [PMID: 26070860 DOI: 10.1586/17474124.2015.1056156] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Liver lipid metabolism and its modulation are involved in many pathologic conditions, such as obesity, non-alcoholic fatty liver disease, diabetes mellitus, atherosclerosis and cardiovascular disease. Metabolic disorders seem to share a similar background of low-grade chronic inflammation, even if the pathophysiological mechanisms leading to tissue and organ damage have not been completely clarified yet. The accumulation of neutral lipids in the liver is now recognized as a beneficial and protective mechanism; on the other hand, lipoperoxidation is involved in the development and progression of non-alcoholic steatohepatitis. The role of the gut microbiota in liver lipid metabolism has been the object of recent scientific investigations. It is likely that the gut microbiota is involved in a complex metabolic modulation and the translocation of gut microflora may also contribute to maintaining the low-grade inflammatory status of metabolic syndrome. Therefore, lipid metabolism pathology has vague limits and complex mechanisms, and the knowledge of these is essential to guide diagnostic and therapeutic decisions.
Collapse
|
26
|
Sobrevia L, Salsoso R, Fuenzalida B, Barros E, Toledo L, Silva L, Pizarro C, Subiabre M, Villalobos R, Araos J, Toledo F, González M, Gutiérrez J, Farías M, Chiarello DI, Pardo F, Leiva A. Insulin Is a Key Modulator of Fetoplacental Endothelium Metabolic Disturbances in Gestational Diabetes Mellitus. Front Physiol 2016; 7:119. [PMID: 27065887 PMCID: PMC4815008 DOI: 10.3389/fphys.2016.00119] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/15/2016] [Indexed: 12/11/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a disease of the mother that associates with altered fetoplacental vascular function. GDM-associated maternal hyperglycaemia result in fetal hyperglycaemia, a condition that leads to fetal hyperinsulinemia and altered L-arginine transport and synthesis of nitric oxide, i.e., endothelial dysfunction. These alterations in the fetoplacental endothelial function are present in women with GDM that were under diet or insulin therapy. Since these women and their newborn show normal glycaemia at term, other factors or conditions could be altered and/or not resolved by restoring normal level of circulating D-glucose. GDM associates with metabolic disturbances, such as abnormal handling of the locally released vasodilator adenosine, and biosynthesis and metabolism of cholesterol lipoproteins, or metabolic diseases resulting in endoplasmic reticulum stress and altered angiogenesis. Insulin acts as a potent modulator of all these phenomena under normal conditions as reported in primary cultures of cells obtained from the human placenta; however, GDM and the role of insulin regarding these alterations in this disease are poorly understood. This review focuses on the potential link between insulin and endoplasmic reticulum stress, hypercholesterolemia, and angiogenesis in GDM in the human fetoplacental vasculature. Based in reports in primary culture placental endothelium we propose that insulin is a factor restoring endothelial function in GDM by reversing ERS, hypercholesterolaemia and angiogenesis to a physiological state involving insulin activation of insulin receptor isoforms and adenosine receptors and metabolism in the human placenta from GDM pregnancies.
Collapse
Affiliation(s)
- Luis Sobrevia
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Faculty of Medicine and Biomedical Sciences, University of Queensland Centre for Clinical Research, University of QueenslandHerston, QLD, Australia; Department of Physiology, Faculty of Pharmacy, Universidad de SevillaSeville, Spain
| | - Rocío Salsoso
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de SevillaSeville, Spain
| | - Bárbara Fuenzalida
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Eric Barros
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Lilian Toledo
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Luis Silva
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Carolina Pizarro
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Mario Subiabre
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Roberto Villalobos
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Joaquín Araos
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío Chillán, Chile
| | - Marcelo González
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de ConcepciónConcepción, Chile; Group of Research and Innovation in Vascular Health (GRIVAS-Health)Chillán, Chile
| | - Jaime Gutiérrez
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Cellular Signaling and Differentiation Laboratory, Health Sciences Faculty, Universidad San SebastiánSantiago, Chile
| | - Marcelo Farías
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Delia I Chiarello
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
27
|
Plasma phospholipid transfer protein (PLTP) modulates adaptive immune functions through alternation of T helper cell polarization. Cell Mol Immunol 2015; 13:795-804. [PMID: 26320740 DOI: 10.1038/cmi.2015.75] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Plasma phospholipid transfer protein (PLTP) is a key determinant of lipoprotein metabolism, and both animal and human studies converge to indicate that PLTP promotes atherogenesis and its thromboembolic complications. Moreover, it has recently been reported that PLTP modulates inflammation and immune responses. Although earlier studies from our group demonstrated that PLTP can modify macrophage activation, the implication of PLTP in the modulation of T-cell-mediated immune responses has never been investigated and was therefore addressed in the present study. Approach and results: In the present study, we demonstrated that PLTP deficiency in mice has a profound effect on CD4+ Th0 cell polarization, with a shift towards the anti-inflammatory Th2 phenotype under both normal and pathological conditions. In a model of contact hypersensitivity, a significantly impaired response to skin sensitization with the hapten-2,4-dinitrofluorobenzene (DNFB) was observed in PLTP-deficient mice compared to wild-type (WT) mice. Interestingly, PLTP deficiency in mice exerted no effect on the counts of total white blood cells, lymphocytes, granulocytes, or monocytes in the peripheral blood. Moreover, PLTP deficiency did not modify the amounts of CD4+ and CD8+ T lymphocyte subsets. However, PLTP-deficiency, associated with upregulation of the Th2 phenotype, was accompanied by a significant decrease in the production of the pro-Th1 cytokine interleukin 18 by accessory cells. CONCLUSIONS For the first time, this work reports a physiological role for PLTP in the polarization of CD4+ T cells toward the pro-inflammatory Th1 phenotype.
Collapse
|
28
|
Deguchi H, Wolfbauer G, Cheung MC, Banerjee Y, Elias DJ, Fernández JA, Albers JJ, Griffin JH. Inhibition of thrombin generation in human plasma by phospholipid transfer protein. Thromb J 2015; 13:24. [PMID: 26185485 PMCID: PMC4504036 DOI: 10.1186/s12959-015-0054-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/20/2015] [Indexed: 01/10/2023] Open
Abstract
Background Plasma phospholipid transfer protein (PLTP) transfers lipids between donors and acceptors (e.g., from HDL to VLDL) and modulates lipoprotein composition, size, and levels. No study has reported an assessment of the effects of PLTP on blood clotting reactions, such as reflected in thrombin generation assays, or on the association of venous thrombosis (VTE) risk with PLTP activity. Methods The in vitro effects of PLTP on blood coagulation reactions and the correlations between plasma PLTP activity levels and VTE were studied. Results Recombinant (r) PLTP concentration-dependently inhibited plasma thrombin generation and factor XII-dependent kallikrein generation when sulfatide was used to stimulate factor XII autoactivation in plasma. However, rPLTP did not inhibit thrombin generation in plasma induced by factor XIa or tissue factor, implicating an effect of PLTP on contact activation reactions. In purified systems, rPLTP inhibited factor XII autoactivation stimulated by sulfatide in the presence of VLDL. In surface plasmon resonance studies, purified factor XII bound to immobilized rPLTP, implying that rPLTP inhibits factor XII-dependent contact activation by binding factor XII in the presence of lipoproteins. Analysis of plasmas from 40 male patients with unprovoked VTE and 40 matched controls indicated that low PLTP lipid transfer activity (≤25th percentile) was associated with an increased risk of VTE after adjustment for body mass index, plasma lipids, and two known thrombophilic genetic risk factors. Conclusion These data imply that PLTP may be an antithrombotic plasma protein by inhibiting generation of prothrombotic factor XIIa in the presence of VLDL. This newly discovered anticoagulant activity of PLTP merits further clinical and biochemical studies. Electronic supplementary material The online version of this article (doi:10.1186/s12959-015-0054-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hiroshi Deguchi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - Gertrud Wolfbauer
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Marian C Cheung
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Yajnavalka Banerjee
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA ; Current Address: Department of Biochemistry, College of Medicine and Health Sciences, SQ University, Muscat, Oman
| | - Darlene J Elias
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - José A Fernández
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - John J Albers
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - John H Griffin
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| |
Collapse
|
29
|
Wahl S, Vogt S, Stückler F, Krumsiek J, Bartel J, Kacprowski T, Schramm K, Carstensen M, Rathmann W, Roden M, Jourdan C, Kangas AJ, Soininen P, Ala-Korpela M, Nöthlings U, Boeing H, Theis FJ, Meisinger C, Waldenberger M, Suhre K, Homuth G, Gieger C, Kastenmüller G, Illig T, Linseisen J, Peters A, Prokisch H, Herder C, Thorand B, Grallert H. Multi-omic signature of body weight change: results from a population-based cohort study. BMC Med 2015; 13:48. [PMID: 25857605 PMCID: PMC4367822 DOI: 10.1186/s12916-015-0282-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/20/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Excess body weight is a major risk factor for cardiometabolic diseases. The complex molecular mechanisms of body weight change-induced metabolic perturbations are not fully understood. Specifically, in-depth molecular characterization of long-term body weight change in the general population is lacking. Here, we pursued a multi-omic approach to comprehensively study metabolic consequences of body weight change during a seven-year follow-up in a large prospective study. METHODS We used data from the population-based Cooperative Health Research in the Region of Augsburg (KORA) S4/F4 cohort. At follow-up (F4), two-platform serum metabolomics and whole blood gene expression measurements were obtained for 1,631 and 689 participants, respectively. Using weighted correlation network analysis, omics data were clustered into modules of closely connected molecules, followed by the formation of a partial correlation network from the modules. Association of the omics modules with previous annual percentage weight change was then determined using linear models. In addition, we performed pathway enrichment analyses, stability analyses, and assessed the relation of the omics modules with clinical traits. RESULTS Four metabolite and two gene expression modules were significantly and stably associated with body weight change (P-values ranging from 1.9 × 10(-4) to 1.2 × 10(-24)). The four metabolite modules covered major branches of metabolism, with VLDL, LDL and large HDL subclasses, triglycerides, branched-chain amino acids and markers of energy metabolism among the main representative molecules. One gene expression module suggests a role of weight change in red blood cell development. The other gene expression module largely overlaps with the lipid-leukocyte (LL) module previously reported to interact with serum metabolites, for which we identify additional co-expressed genes. The omics modules were interrelated and showed cross-sectional associations with clinical traits. Moreover, weight gain and weight loss showed largely opposing associations with the omics modules. CONCLUSIONS Long-term weight change in the general population globally associates with serum metabolite concentrations. An integrated metabolomics and transcriptomics approach improved the understanding of molecular mechanisms underlying the association of weight gain with changes in lipid and amino acid metabolism, insulin sensitivity, mitochondrial function as well as blood cell development and function.
Collapse
|
30
|
Elevated baseline plasma phospholipid protein (PLTP) levels are an independent predictor of long-term all-cause mortality in patients with diabetes mellitus and known or suspected coronary artery disease. Atherosclerosis 2015; 239:503-8. [PMID: 25710294 DOI: 10.1016/j.atherosclerosis.2015.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/11/2015] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To investigate the long-term prognostic significance of baseline plasma PLTP levels in a group of well-characterized male patients with diabetes mellitus and known or suspected coronary artery disease referred for coronary angiography. BACKGROUND PLTP is a plasma protein that mediates the net transfer and exchange of phospholipids between lipoproteins. It has been implicated in the pathogenesis of atherosclerosis and elevated plasma levels have been reported in patients with diabetes mellitus. METHODS Baseline plasma PLTP levels were measured in 154 male patients with diabetes mellitus who were referred for coronary angiography and followed prospectively for 5 years for the development of all-cause mortality. RESULTS After adjustment for a variety of baseline clinical, angiographic and laboratory parameters, plasma PLTP levels (analyzed as a continuous variable) were an independent predictor of all-cause mortality at 5 years (HR, 1.55; 95% CI, 1.22-2.00; P = 0.0009). Furthermore, in 3 additional multivariate models that also included a wide variety of contemporary biomarkers with established prognostic efficacy (i.e., ST2, GDF-15, Cystatin C, Fibrinogen, and NT-proBNP), PLTP remained an independent predictor of all-cause mortality at 5 years. CONCLUSIONS Elevated baseline plasma levels of PLTP are associated with an increased risk of long-term all-cause mortality in patients with diabetes and known or suspected coronary disease. Furthermore, this association is independent of a variety of clinical, angiographic, and laboratory variables, including a whole host of contemporary biomarkers with established prognostic efficacy.
Collapse
|
31
|
Abbasi A, Dallinga-Thie GM, Dullaart RP. Phospholipid transfer protein activity and incident type 2 diabetes mellitus. Clin Chim Acta 2015; 439:38-41. [DOI: 10.1016/j.cca.2014.09.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 11/25/2022]
|
32
|
Chirackal Manavalan AP, Kober A, Metso J, Lang I, Becker T, Hasslitzer K, Zandl M, Fanaee-Danesh E, Pippal JB, Sachdev V, Kratky D, Stefulj J, Jauhiainen M, Panzenboeck U. Phospholipid transfer protein is expressed in cerebrovascular endothelial cells and involved in high density lipoprotein biogenesis and remodeling at the blood-brain barrier. J Biol Chem 2014; 289:4683-98. [PMID: 24369175 PMCID: PMC3931031 DOI: 10.1074/jbc.m113.499129] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Phospholipid transfer protein (PLTP) is a key protein involved in biogenesis and remodeling of plasma HDL. Several neuroprotective properties have been ascribed to HDL. We reported earlier that liver X receptor (LXR) activation promotes cellular cholesterol efflux and formation of HDL-like particles in an established in vitro model of the blood-brain barrier (BBB) consisting of primary porcine brain capillary endothelial cells (pBCEC). Here, we report PLTP synthesis, regulation, and its key role in HDL metabolism at the BBB. We demonstrate that PLTP is highly expressed and secreted by pBCEC. In a polarized in vitro model mimicking the BBB, pBCEC secreted phospholipid-transfer active PLTP preferentially to the basolateral ("brain parenchymal") compartment. PLTP expression levels and phospholipid transfer activity were enhanced (up to 2.5-fold) by LXR activation using 24(S)-hydroxycholesterol (a cerebral cholesterol metabolite) or TO901317 (a synthetic LXR agonist). TO901317 administration elevated PLTP activity in BCEC from C57/BL6 mice. Preincubation of HDL3 with human plasma-derived active PLTP resulted in the formation of smaller and larger HDL particles and enhanced the capacity of the generated HDL particles to remove cholesterol from pBCEC by up to 3-fold. Pre-β-HDL, detected by two-dimensional crossed immunoelectrophoresis, was generated from HDL3 in pBCEC-derived supernatants, and their generation was markedly enhanced (1.9-fold) upon LXR activation. Furthermore, RNA interference-mediated PLTP silencing (up to 75%) reduced both apoA-I-dependent (67%) and HDL3-dependent (30%) cholesterol efflux from pBCEC. Based on these findings, we propose that PLTP is actively involved in lipid transfer, cholesterol efflux, HDL genesis, and remodeling at the BBB.
Collapse
Affiliation(s)
| | | | - Jari Metso
- the National Institute for Health and Welfare, Biomedicum, FI-00290 Helsinki, Finland, and
| | - Ingrid Lang
- Institute of Cell Biology, Histology, and Embryology, and
| | | | | | - Martina Zandl
- From the Institute of Pathophysiology and Immunology
| | | | | | - Vinay Sachdev
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Jasminka Stefulj
- the Department of Molecular Biology, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Matti Jauhiainen
- the National Institute for Health and Welfare, Biomedicum, FI-00290 Helsinki, Finland, and
| | - Ute Panzenboeck
- From the Institute of Pathophysiology and Immunology, , To whom correspondence should be addressed: Institute of Pathophysiology and Immunology, Medical University of Graz, Heinrichstrasse 31a, 8010 Graz, Austria. Tel.: 43-316-3801955; Fax: 43-316-3809640; E-mail:
| |
Collapse
|
33
|
Abstract
Cardiovascular disease represents the most common cause of death in patients with nonalcoholic fatty liver disease (NAFLD). Patients with NAFLD exhibit an atherogenic dyslipidemia that is characterized by an increased plasma concentration of triglycerides, reduced concentration of high-density lipoprotein (HDL) cholesterol, and low-density lipoprotein (LDL) particles that are smaller and more dense than normal. The pathogenesis of NAFLD-associated atherogenic dyslipidemia is multifaceted, but many aspects are attributable to manifestations of insulin resistance. Here the authors review the structure, function, and metabolism of lipoproteins, which are macromolecular particles of lipids and proteins that transport otherwise insoluble triglyceride and cholesterol molecules within the plasma. They provide a current explanation of the metabolic perturbations that are observed in the setting of insulin resistance. An improved understanding of the pathophysiology of atherogenic dyslipidemia would be expected to guide therapies aimed at reducing morbidity and mortality in patients with NAFLD.
Collapse
Affiliation(s)
- Edward Fisher
- Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York
| | - David Cohen
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Deckert V, Kretz B, Habbout A, Raghay K, Labbé J, Abello N, Desrumaux C, Gautier T, Lemaire-Ewing S, Maquart G, Le Guern N, Masson D, Steinmetz E, Lagrost L. Development of abdominal aortic aneurysm is decreased in mice with plasma phospholipid transfer protein deficiency. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:975-86. [PMID: 23830874 DOI: 10.1016/j.ajpath.2013.05.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 04/12/2013] [Accepted: 05/13/2013] [Indexed: 12/13/2022]
Abstract
Plasma phospholipid transfer protein (PLTP) increases the circulating levels of proatherogenic lipoproteins, accelerates blood coagulation, and modulates inflammation. The role of PLTP in the development of abdominal aortic aneurysm (AAA) was investigated by using either a combination of mechanical and elastase injury at one site of mouse aorta (elastase model) or continuous infusion of angiotensin II in hyperlipidemic ApoE-knockout mice (Ang II model). With the elastase model, complete PLTP deficiency was associated with a significantly lower incidence and a lesser degree of AAA expansion. With the Ang II model, findings were consistent with those in the elastase model, with a lower severity grade in PLTP-deficient mice, an intermediate phenotype in PLTP-deficient heterozygotes, and a blunted effect of the PLTP-deficient trait when restricted to bone marrow-derived immune cells. The protective effect of whole-body PLTP deficiency in AAA was illustrated further by a lesser degree of adventitia expansion, reduced elastin degradation, fewer recruited macrophages, and less smooth muscle cell depletion in PLTP-deficient than in wild-type mice, as evident from comparative microscopic analysis of aorta sections. Finally, cumulative evidence supports the association of PLTP deficiency with reduced expression and activity levels of matrix metalloproteinases, known to degrade elastin and collagen. We conclude that PLTP can play a significant role in the pathophysiology of AAA.
Collapse
|
35
|
Phospholipid transfer protein, an emerging cardiometabolic risk marker: Is it time to intervene? Atherosclerosis 2013; 228:38-41. [DOI: 10.1016/j.atherosclerosis.2013.01.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/13/2022]
|
36
|
Desrumaux C, Pisoni A, Meunier J, Deckert V, Athias A, Perrier V, Villard V, Lagrost L, Verdier JM, Maurice T. Increased amyloid-β peptide-induced memory deficits in phospholipid transfer protein (PLTP) gene knockout mice. Neuropsychopharmacology 2013; 38:817-25. [PMID: 23303044 PMCID: PMC3671992 DOI: 10.1038/npp.2012.247] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Oxidative stress is recognized as one of the earliest and most intense pathological processes in Alzheimer's disease (AD), and the antioxidant vitamin E has been shown to efficiently prevent amyloid plaque formation and neurodegeneration. Plasma phospholipid transfer protein (PLTP) has a major role in vitamin E transfers in vivo, and PLTP deficiency in mice is associated with reduced brain vitamin E levels. To determine the impact of PLTP on amyloid pathology in vivo, we analyzed the vulnerability of PLTP-deficient (PLTP-KO) mice to the toxic effects induced by intracerebroventricular injection of oligomeric amyloid-β 25-35 (Aβ 25-35) peptide, a non-transgenic model of AD. Under basal conditions, PLTP-KO mice showed increased cerebral oxidative stress, increased brain Aβ 1-42 levels, and a lower expression of the synaptic function marker synaptophysin, as compared with wild-type mice. This PLTP-KO phenotype was associated with increased memory impairment 1 week after Aβ25-35 peptide injection. Restoration of brain vitamin E levels in PLTP-KO mice through a chronic dietary supplementation prevented Aβ 25-35-induced memory deficits and reduced cerebral oxidative stress and toxicity. We conclude that PLTP, through its ability to deliver vitamin E to the brain, constitutes an endogenous neuroprotective agent. Increasing PLTP activity may offer a new way to develop neuroprotective therapies.
Collapse
Affiliation(s)
- Catherine Desrumaux
- INSERM U710, Université Montpellier 2, CC105, Place Eugène Bataillon, 34095 Montpellier, Cedex 05, France.
| | - Amandine Pisoni
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France
| | | | | | - Anne Athias
- Lipidomics analytical platform, SFR 100, Dijon, France
| | - Véronique Perrier
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France
| | | | | | - Jean-Michel Verdier
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France
| | - Tangui Maurice
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France,INSERM U710, Université Montpellier 2, CC105, Place Eugène Bataillon, 34095 Montpellier, Cedex 05, France. Tel: (+33/0) 4 67 14 36 23, Fax: (+33/0) 4 67 14 92 95, E-mail: or E-mail:
| |
Collapse
|
37
|
Abstract
Cardiac and peripheral vascular biomarkers are increasingly becoming targets of both research and clinical practice. As of 2008, cardiovascular-related medical care accounts for greater than 20% of all the economic costs of illness in the United States. In the age of burgeoning financial pressures on the entire health care system, never has it been more important to try to understand who is at risk for cardiovascular disease in order to prevent new events. In this paper, we will discuss the cost of cardiovascular disease to society, clarify the definition of and need for biomarkers, offer an example of a current biomarker, namely high-sensitivity C-reactive protein, and finally examine the approval process for utilizing these in clinical practice.
Collapse
Affiliation(s)
- Jana E Montgomery
- Dartmouth-Hitchcock Medical Center, Division of Cardiology, Lebanon, NH 03756, USA.
| | | |
Collapse
|
38
|
Lin HY, Yin Y, Zhang JX, Xuan H, Zheng Y, Zhan SS, Zhu YX, Han X. Identification of direct forkhead box O1 targets involved in palmitate-induced apoptosis in clonal insulin-secreting cells using chromatin immunoprecipitation coupled to DNA selection and ligation. Diabetologia 2012; 55:2703-2712. [PMID: 22810813 DOI: 10.1007/s00125-012-2643-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 06/11/2012] [Indexed: 01/10/2023]
Abstract
AIMS/HYPOTHESIS The transcription factor, forkhead box (FOX)O1, is involved in fatty acid-induced apoptosis in pancreatic beta cells, but the precise mechanism is poorly understood. We aimed to identify which direct downstream targets of FOXO1 are involved in palmitate-induced apoptosis in the pancreatic beta cell line MIN6. METHODS Chromatin immunoprecipitation (ChIP) coupled to a DNA selection and ligation technique (ChIP-DSL) was used to identify the direct targets of FOXO1. The mRNA level was examined by real-time PCR assay. The ChIP-DSL results were verified using ChIP-PCR and luciferase assay, respectively. The cell apoptosis rate was determined by TUNEL assay and by scoring cells with pycnotic nuclei. RESULTS We identified 189 target genes and selected 106 targets for expression analysis in MIN6 cells treated with palmitate. The results showed that six genes were significantly upregulated and four were downregulated. Binding of FOXO1 to the promoters was determined by ChIP-PCR and confirmed by luciferase assay. Among the ten up- and downregulated genes, mRNA expression of A930038C07Rik was significantly decreased and that of Ppa1 was increased in 8-week-old db/db mice. The apoptosis assay showed that overproduction of the protein 'RIKEN cDNA A930038C07' (A930038C07Rik) drastically enhanced palmitate-induced apoptosis, while pyrophosphatase (inorganic) 1 (PPA1) partially protected the cells from apoptosis. Knockdown of PPA1, moreover, significantly increased apoptosis. CONCLUSIONS/INTERPRETATION We identified for the first time FOXO1 targets in MIN6 cells treated with palmitate, thus revealing the important roles of A930038C07Rik and PPA1 in palmitate-induced cell apoptosis. These results shed light on the mechanisms of palmitate-induced apoptosis in pancreatic beta cells.
Collapse
Affiliation(s)
- H Y Lin
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China
| | - Y Yin
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China
| | - J X Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China
| | - H Xuan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China
| | - Y Zheng
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China
| | - S S Zhan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China
| | - Y X Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China
| | - X Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
39
|
Zhang L, Michal JJ, O'Fallon JV, Pan Z, Gaskins CT, Reeves JJ, Busboom JR, Zhou X, Ding B, Dodson MV, Jiang Z. Quantitative genomics of 30 complex phenotypes in Wagyu x Angus F₁ progeny. Int J Biol Sci 2012; 8:838-58. [PMID: 22745575 PMCID: PMC3385007 DOI: 10.7150/ijbs.4403] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 06/04/2012] [Indexed: 12/25/2022] Open
Abstract
In the present study, a total of 91 genes involved in various pathways were investigated for their associations with six carcass traits and twenty-four fatty acid composition phenotypes in a Wagyu×Angus reference population, including 43 Wagyu bulls and their potential 791 F1 progeny. Of the 182 SNPs evaluated, 102 SNPs that were in Hardy-Weinberg equilibrium with minor allele frequencies (MAF>0.15) were selected for parentage assignment and association studies with these quantitative traits. The parentage assignment revealed that 40 of 43 Wagyu sires produced over 96.71% of the calves in the population. Linkage disequilibrium analysis identified 75 of 102 SNPs derived from 54 genes as tagged SNPs. After Bonferroni correction, single-marker analysis revealed a total of 113 significant associations between 44 genes and 29 phenotypes (adjusted P<0.05). Multiple-marker analysis confirmed single-gene associations for 10 traits, but revealed two-gene networks for 9 traits and three-gene networks for 8 traits. Particularly, we observed that TNF (tumor necrosis factor) gene is significantly associated with both beef marbling score (P=0.0016) and palmitic acid (C16:0) (P=0.0043), RCAN1 (regulator of calcineurin 1) with rib-eye area (P=0.0103), ASB3 (ankyrin repeat and SOCS box-containing 3) with backfat (P=0.0392), ABCA1 (ATP-binding cassette A1) with both palmitic acid (C16:0) (P=0.0025) and oleic acid (C18:1n9) (P=0.0114), SLC27A1(solute carrier family 27 A1) with oleic acid (C18:1n9) (P=0.0155), CRH (corticotropin releasing hormone) with both linolenic acid (OMEGA-3) (P=0.0200) and OMEGA 6:3 RATIO (P=0.0054), SLC27A2 (solute carrier family 27 A2) with both linoleic acid (OMEGA-6) (P=0.0121) and FAT (P=0.0333), GNG3 (guanine nucleotide binding protein gamma 3 with desaturase 9 (P=0.0115), and EFEMP1 (EGF containing fibulin-like extracellular matrix protein 1), PLTP (phospholipid transfer protein) and DSEL (dermatan sulfate epimerase-like) with conjugated linoleic acid (P=0.0042-0.0044), respectively, in the Wagyu x Angus F1 population. In addition, we observed an interesting phenomenon that crossbreeding of different breeds might change gene actions to dominant and overdominant modes, thus explaining the origin of heterosis. The present study confirmed that these important families or pathway-based genes are useful targets for improving meat quality traits and healthful beef products in cattle.
Collapse
Affiliation(s)
- Lifan Zhang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164-6351, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Vaisberg M, Bachi ALL, Latrilha C, Dioguardi GS, Bydlowski SP, Maranhão RC. Lipid transfer to HDL is higher in marathon runners than in sedentary subjects, but is acutely inhibited during the run. Lipids 2012; 47:679-86. [PMID: 22684913 DOI: 10.1007/s11745-012-3685-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 05/17/2012] [Indexed: 12/21/2022]
Abstract
Although exercise increases HDL-cholesterol, exercise-induced changes in HDL metabolism have been little explored. Lipid transfer to HDL is essential for HDL's role in reverse cholesterol transport. We investigated the effects of acute exhaustive exercise on lipid transfer to HDL. We compared plasma lipid, apolipoprotein and cytokine levels and in vitro transfer of four lipids from a radioactively labeled lipid donor nanoemulsion to HDL in sedentary individuals (n = 28) and in marathon runners (n = 14) at baseline, immediately after and 72 h after a marathon. While HDL-cholesterol concentrations and apo A1 levels were higher in marathon runners, LDL-cholesterol, apo B and triacylglycerol levels were similar in both groups. Transfers of non-esterified cholesterol [6.8 (5.7-7.2) vs. 5.2 (4.5-6), p = 0.001], phospholipids [21.7 (20.4-22.2) vs. 8.2 (7.7-8.9), p = 0.0001] and triacylglycerol [3.7 (3.1-4) vs. 1.3 (0.8-1.7), p = 0.0001] were higher in marathon runners, but esterified-cholesterol transfer was similar. Immediately after the marathon, LDL- and HDL-cholesterol concentrations and apo A1 levels were unchanged, but apo B and triacylglycerol levels increased. Lipid transfer of non-esterified cholesterol [6.8 (5.7-7.2) vs. 5.8 (4.9-6.6), p = 0.0001], phospholipids [21.7 (20.4-22.2) vs. 19.1 (18.6-19.3), p = 0.0001], esterified-cholesterol [3.2 (2.2-3.8) vs. 2.3 (2-2.9), p = 0.02] and triacylglycerol [3.7 (3.1-4) vs. 2.6 (2.1-2.8), p = 0.0001] to HDL were all reduced immediately after the marathon but returned to baseline 72 h later. Running a marathon increased IL-6 and TNF-α levels, but after 72 h these values returned to baseline. Lipid transfer, except esterified-cholesterol transfer, was higher in marathon runners than in sedentary individuals, but the marathon itself acutely inhibited lipid transfer. In light of these novel observations, further study is required to clarify how these metabolic changes can influence HDL composition and anti-atherogenic function.
Collapse
Affiliation(s)
- Mauro Vaisberg
- Department of Otorhinolaryngology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
41
|
Dullaart RPF, Vergeer M, de Vries R, Kappelle PJWH, Dallinga-Thie GM. Type 2 diabetes mellitus interacts with obesity and common variations in PLTP to affect plasma phospholipid transfer protein activity. J Intern Med 2012; 271:490-8. [PMID: 21973210 DOI: 10.1111/j.1365-2796.2011.02465.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Phospholipid transfer protein (PLTP) is an emerging cardiometabolic risk marker that is important in high-density lipoprotein (HDL) and triglyceride metabolism. Plasma PLTP activity is elevated in type 2 diabetes mellitus, whereas glucose may regulate PLTP gene transcription in vitro. Of interest, common PLTP variations that predict cardiovascular disease have been identified recently. We investigated whether the diabetic state is able to amplify relationships between obesity and PLTP gene variations with circulating PLTP levels. SUBJECTS AND METHODS Plasma PLTP activity (using a phospholipid vesicles-HDL system), PLTP gene score [number of PLTP activity-decreasing alleles based on two tagging polymorphisms (rs378114 and rs60- 65904)] and waist circumference were determined in two Dutch cohorts comprising 237 patients with type 2 diabetes and 78 control subjects. RESULTS Patients with diabetes were more obese (P < 0.001 for prevalence of increased waist circumference) and had 13% higher plasma PLTP activity (P < 0.001). PLTP gene score was not different in diabetic and control subjects (P = 0.40). PLTP activity was highest in patients with diabetes with an enlarged waist and lowest in control subjects with a normal waist circumference (P < 0.001). Multiple linear regression analysis revealed a positive interaction between diabetes status and waist circumference on PLTP activity (β = 0.200, P = 0.005). Furthermore, diabetes status (β = -0.485, P = 0.046) or HbA1c (β = -0.240, P = 0.035) interacted with PLTP gene score to affect PLTP activity. CONCLUSIONS Type 2 diabetes and enlarged waist circumference interact to impact on plasma PLTP activity. Diabetes may also amplify the association between plasma PLTP activity and common PLTP gene variations. Our findings support the hypothesis that diabetes-environment and diabetes-gene interactions govern plasma PLTP activity.
Collapse
Affiliation(s)
- R P F Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
42
|
Al-Zoairy R, Melmer A, Ress C, Laimer M, Kaser S, Ebenbichler C. Lipid profile changes after pronounced weight loss induced by bariatric surgery. ACTA ACUST UNITED AC 2012. [DOI: 10.2217/clp.12.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
43
|
Côté M, Provost PR, Tremblay Y. Apolipoprotein A-I, A-II, and H mRNA and protein accumulation sites in the developing lung in late gestation. BMC Res Notes 2011; 4:235. [PMID: 21756353 PMCID: PMC3154161 DOI: 10.1186/1756-0500-4-235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 07/14/2011] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Expression of apolipoprotein A-I (apoA-I), A-II, and H was previously observed at 16 to 50-fold higher levels in the fetal than the adult mouse lung. Here, sites of apoA-I, A-II, and H mRNA and protein accumulation were determined in mouse fetal lungs by in situ hybridization and immunohistochemistry in late gestation. RESULTS Expression sites vary for the three genes and change for the distal epithelium before the end of the canalicular stage, thus where and when the surge of surfactant synthesis occurs. Messenger of apoH, but not those of apoA-I and A-II, was also observed in the proximal epithelium and smooth muscles surrounding arteries. In contrast to apoC-II protein, none of the three studied apolipoproteins accumulated within secretory granule-like structures. Immunohistochemistry revealed that apoA-I and apoH accumulated mainly in capillaries. Three different positive signals with the anti-apoA-II antibody were found: one transient signal in the nucleus of a portion of mesenchymal cells, a second at lower levels throughout the mesenchyme, and another in capillaries with a specific increase from gestation day 17.5/18.5. CONCLUSION Temporal and geographic co-expression of apoAI, AII, and H genes with surfactant production site suggests that the three apolipoproteins are secreted to play roles supporting the lung-specific surfactant lipid-related metabolism.
Collapse
Affiliation(s)
- Mélissa Côté
- Reproduction Axis, Perinatal and Child Health, Rm T-1-49, CHUQ Research Center, Québec City, Québec, Canada.
| | | | | |
Collapse
|
44
|
de Vries R, Kappelle PJ, Dallinga-Thie GM, Dullaart RP. Plasma phospholipid transfer protein activity is independently determined by obesity and insulin resistance in non-diabetic subjects. Atherosclerosis 2011; 217:253-9. [DOI: 10.1016/j.atherosclerosis.2011.03.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 03/03/2011] [Accepted: 03/14/2011] [Indexed: 12/13/2022]
|
45
|
Tailleux A, Staels B. Overview of the Measurement of Lipids and Lipoproteins in Mice. ACTA ACUST UNITED AC 2011; 1:265-77. [DOI: 10.1002/9780470942390.mo110001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Anne Tailleux
- Université Lille Nord de France Lille France
- Inserm, U1011 Lille France
- UDSL Lille France
- Institut Pasteur de Lille Lille France
| | - Bart Staels
- Université Lille Nord de France Lille France
- Inserm, U1011 Lille France
- UDSL Lille France
- Institut Pasteur de Lille Lille France
| |
Collapse
|
46
|
Azevedo CHM, Wajngarten M, Prete ACL, Diament J, Maranhão RC. Simultaneous transfer of cholesterol, triglycerides, and phospholipids to high-density lipoprotein in aging subjects with or without coronary artery disease. Clinics (Sao Paulo) 2011; 66:1543-8. [PMID: 22179156 PMCID: PMC3164401 DOI: 10.1590/s1807-59322011000900006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 05/16/2011] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To verify whether the capacity of high-density lipoprotein (HDL) to simultaneously receive nonesterified cholesterol, triglycerides, cholesteryl esters, and phospholipids changes with aging and the presence of coronary artery disease. DESIGN Cross-sectional study with biochemical analyses. SUBJECTS Eleven elderly patients with coronary artery disease (74 ± 5 years) were compared with the following groups of non-coronary artery disease subjects (referred to as "healthy"): 25 young (25 ± 5 years), 25 middle-aged (42 ± 6 years), and 25 elderly subjects (75 ± 8 years). METHODS Plasma samples were incubated with a nanoemulsion labeled with radioactive lipids; the transfer of the lipids from the nanoemulsion to the HDL was measured in chemically precipitated HDL. HDL size and paraoxonase-1 activity were also determined. RESULTS The transfer of cholesteryl esters and phospholipids to high-density lipoprotein was significantly greater (p<0.001) in healthy elderly subjects than in the middle-aged and younger subjects. Non-esterified cholesterol and triglyceride transfer was not different among these three groups. The HDL size was significantly greater (p<0.001) in healthy elderly subjects than in the middle-aged and younger subjects. The paraoxonase-1 activity was similar among the groups. Compared with healthy elderly subjects, coronary artery disease elderly subjects had significantly less (p<0.05) transfer of non-esterified cholesterol, triglycerides, and cholesteryl esters to the HDL and a significantly smaller (p<0.05) HDL size. CONCLUSION Because lipid transfer is enhanced in healthy elderly subjects but not in those with coronary artery disease, increasing lipid transfer to HDL may be a protective mechanism against the disease.
Collapse
Affiliation(s)
- Carolina H M Azevedo
- Lipid Metabolism Laboratory, Heart Institute, Faculdade de Medicina da Universidade de São Paulo, Brazil
| | | | | | | | | |
Collapse
|
47
|
Affiliation(s)
- William R Lagor
- Institute for Translational Medicine and Therapeutics, and Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Daniel J Rader
- Institute for Translational Medicine and Therapeutics, and Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
48
|
Desrumaux C, Deckert V, Lemaire-Ewing S, Mossiat C, Athias A, Vandroux D, Dumont L, Monier S, Pais de Barros JP, Klein A, De Maistre E, Blache D, Beley A, Marie C, Garnier P, Lagrost L. Plasma phospholipid transfer protein deficiency in mice is associated with a reduced thrombotic response to acute intravascular oxidative stress. Arterioscler Thromb Vasc Biol 2010; 30:2452-7. [PMID: 20864671 DOI: 10.1161/atvbaha.110.207654] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Earlier in vitro studies suggested a putative role for the plasma phospholipid transfer protein (PLTP) in the modulation of blood coagulation. The effect of PLTP expression on blood coagulation under both basal and oxidative stress conditions was compared here in wild-type and PLTP-deficient (PLTP-/-) mice. METHODS AND RESULTS Under basal conditions, PLTP deficiency was associated with an extended tail bleeding time despite a significant depletion of vascular α-tocopherol content and an impairment of endothelial function. When acute oxidative stress was generated in vivo in the brain vasculature, the steady state levels of oxidized lipid derivatives, the extent of blood vessel occlusion, and the volume of ischemic lesions were more severe in wild-type than in PLTP-/- mice. CONCLUSIONS In addition to its recognized hyperlipidemic, proinflammatory, and proatherogenic properties, PLTP increases blood coagulation and worsens the extent of ischemic lesions in response to acute oxidative stress. Thus, PLTP arises here as a cardiovascular risk factor for the late thrombotic events occurring in the acute phase of atherosclerosis.
Collapse
Affiliation(s)
- Catherine Desrumaux
- Institut National de la Santé et de la Recherche Médicale, UMR866, Dijon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lemaire-Ewing S, Desrumaux C, Néel D, Lagrost L. Vitamin E transport, membrane incorporation and cell metabolism: Is alpha-tocopherol in lipid rafts an oar in the lifeboat? Mol Nutr Food Res 2010; 54:631-40. [PMID: 20166147 DOI: 10.1002/mnfr.200900445] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Vitamin E is composed of closely related compounds, including tocopherols and tocotrienols. Studies of the last decade provide strong support for a specific role of alpha-tocopherol in cell signalling and the regulation of gene expression. It produces significant effects on inflammation, cell proliferation and apoptosis that are not shared by other vitamin E isomers with similar antioxidant properties. The different behaviours of vitamin E isomers might relate, at least in part, to the specific effects they exert at the plasma membrane. alpha-Tocopherol is not randomly distributed throughout the phospholipid bilayer of biological membranes, and as compared with other isomers, it shows a propensity to associate with lipid rafts. Distinct aspects of vitamin E transport and metabolism is discussed with emphasis on the interaction between alpha-tocopherol and lipid rafts and the consequences of these interactions on cell metabolism.
Collapse
|
50
|
Saunders RA, Fujii K, Alabanza L, Ravatn R, Kita T, Kudoh K, Oka M, Chin KV. Altered phospholipid transfer protein gene expression and serum lipid profile by topotecan. Biochem Pharmacol 2010; 80:362-9. [PMID: 20416282 PMCID: PMC2883626 DOI: 10.1016/j.bcp.2010.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 04/11/2010] [Accepted: 04/13/2010] [Indexed: 01/19/2023]
Abstract
Camptothecin (CPT) and its structural analogues including topotecan and irinotecan, are inhibitors of topoisomerase I. These drugs are clinically active against a broad spectrum of cancers. To understand the genesis of chemotherapeutic resistance to the CPT family of anticancer drugs, we examined by gene expression profiling the pharmacological response to topotecan in the human hepatoma HepG2 cells and found a striking induction of the phospholipid transfer protein (PLTP) gene expression by topotecan. We showed that activation of PLTP gene expression is specific to CPT and its analogues including specific enantiomers that inhibit topoisomerase I. PLTP-mediated lipid transfer to high-density lipoprotein (HDL) is thought to be important for shuttling and redistribution of lipids between lipoproteins, which are normally returned to the liver for metabolism via the reverse cholesterol transport pathway. Hence, we asked whether elevated PLTP levels might increase the transfer of drugs into HDL. We observed that CPT was not accumulated in HDL and other lipoproteins. In addition, topotecan treatment in mice caused a marked reduction in serum HDL that was accompanied by an increase in triglyceride and cholesterol levels. These results showed that PLTP does not mediate the transfer of topoisomerase I inhibitors to serum lipoproteins. However, elevated serum PLTP levels following treatment with topoisomerase I inhibitors in cancer patients may serve as a biomarker for monitoring the development of hypertriglyceridemia and acute pancreatitis.
Collapse
Affiliation(s)
- Rudel A. Saunders
- Department of Medicine, The University of Toledo, College of Medicine, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, The University of Toledo, College of Medicine, Toledo, OH United States
| | - Kazuyuki Fujii
- Department of Medicine, The University of Toledo, College of Medicine, Toledo, OH, United States
- Department of Obstetrics and Gynecology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Leah Alabanza
- Department of Medicine, The University of Toledo, College of Medicine, Toledo, OH, United States
- Baker Institute for Animal Health, Cornell Veterinary College, Ithaca, NY, United States
| | - Roald Ravatn
- Department of Medicine, The University of Toledo, College of Medicine, Toledo, OH, United States
| | - Tsunekazu Kita
- Department of Gynecology, Saitama Cancer Center, Adachi-Gun, Japan
| | - Kazuya Kudoh
- Department of Obstetrics and Gynecology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Masahiro Oka
- Division of Dermatology, Department of Clinical Molecular Medicine, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Khew-Voon Chin
- Department of Medicine, The University of Toledo, College of Medicine, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, The University of Toledo, College of Medicine, Toledo, OH United States
| |
Collapse
|