1
|
Spanu D, Pretta A, Lai E, Persano M, Donisi C, Mariani S, Dubois M, Migliari M, Saba G, Ziranu P, Pusceddu V, Puzzoni M, Astara G, Scartozzi M. Hepatocellular carcinoma and microbiota: Implications for clinical management and treatment. World J Hepatol 2022; 14:1319-1332. [PMID: 36158925 PMCID: PMC9376771 DOI: 10.4254/wjh.v14.i7.1319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota plays an essential role in host homeostasis. It is involved in several physiological processes such as nutrients digestion and absorption, maintenance of intestinal epithelial barrier integrity and immune system self-tolerance. Especially the gut microbiota is assumed to play a crucial role in many gastrointestinal, pancreatic and liver disorders. Its role in hepatic carcinogenesis is also gaining increasing interest, especially regarding the development of therapeutic strategies. Different studies are highlighting a link between some bacterial strains and liver disease, including hepatocellular carcinoma (HCC). Indeed, HCC represents an interesting field of research in this perspective, due to the gut-liver axis, to the implication of microbiota in the immune system and to the increasing number of immunotherapy agents investigated in this tumour. Thus, the assessment of the role of microbiota in influencing clinical outcome for patients treated with these drugs is becoming of increasing importance. Our review aims to give an overview on the relationship between microbiota and HCC development/progression and treatment. We focus on potential implications on the available treatment strategies and those under study in the various stages of disease. We highlight the pathogenic mechanisms and investigate the underlying molecular pathways involved. Moreover, we investigate the potential prognostic and/or predictive role of microbiota for target therapies, immune checkpoint inhibitors and loco-regional treatment. Finally, given the limitation of current treatments, we analyze the gut microbiota-mediated therapies and its potential options for HCC treatment focusing on fecal microbiota transplantation.
Collapse
Affiliation(s)
- Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Mara Persano
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Clelia Donisi
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Marco Dubois
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Marco Migliari
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Giorgio Astara
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, Monserrato 09042, Cagliari, Italy
| |
Collapse
|
2
|
Sphingolipids and Cholesterol. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:1-14. [DOI: 10.1007/978-981-19-0394-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
3
|
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
|
4
|
Nonalcoholic Fatty Liver Disease (NAFLD) as Model of Gut-Liver Axis Interaction: From Pathophysiology to Potential Target of Treatment for Personalized Therapy. Int J Mol Sci 2021; 22:ijms22126485. [PMID: 34204274 PMCID: PMC8233936 DOI: 10.3390/ijms22126485] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading cause of liver disease worldwide, affecting both adults and children and will result, in the near future, as the leading cause of end-stage liver disease. Indeed, its prevalence is rapidly increasing, and NAFLD is becoming a major public health concern. For this reason, great efforts are needed to identify its pathogenetic factors and new therapeutic approaches. In the past decade, enormous advances understanding the gut-liver axis-the complex network of cross-talking between the gut, microbiome and liver through the portal circulation-have elucidated its role as one of the main actors in the pathogenesis of NAFLD. Indeed, evidence shows that gut microbiota is involved in the development and progression of liver steatosis, inflammation and fibrosis seen in the context of NAFLD, as well as in the process of hepatocarcinogenesis. As a result, gut microbiota is currently emerging as a non-invasive biomarker for the diagnosis of disease and for the assessment of its severity. Additionally, to its enormous diagnostic potential, gut microbiota is currently studied as a therapeutic target in NAFLD: several different approaches targeting the gut homeostasis such as antibiotics, prebiotics, probiotics, symbiotics, adsorbents, bariatric surgery and fecal microbiota transplantation are emerging as promising therapeutic options.
Collapse
|
5
|
Li Z, Chiang YP, He M, Zhang K, Zheng J, Wu W, Cai J, Chen Y, Chen G, Chen Y, Dong J, Worgall TS, Jiang XC. Effect of liver total sphingomyelin synthase deficiency on plasma lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158898. [PMID: 33545384 DOI: 10.1016/j.bbalip.2021.158898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 11/26/2022]
Abstract
Sphingomyelin (SM) is one major phospholipids on lipoproteins. It is enriched on apolipoprotein B-containing particles, including very low-density lipoprotein (VLDL) and its catabolites, low-density lipoprotein (LDL). SM is synthesized by sphingomyelin synthase 1 and 2 (SMS1 and SMS2) which utilizes ceramide and phosphatidylcholine, as two substrates, to produce SM and diacylglyceride. SMS1 and SMS2 activities are co-expressed in all tested tissues, including the liver where VLDL is produced. Thus, neither Sms1 gene knockout (KO) nor Sms2 KO approach is sufficient to evaluate the effect of SMS on VLDL metabolism. We prepared liver-specific Sms1 KO/global Sms2 KO mice to evaluate the effect of hepatocyte SM biosynthesis in lipoprotein metabolism. We found that hepatocyte total SMS depletion significantly reduces cellular sphingomyelin levels. Also, we found that the deficiency induces cellular glycosphingolipid levels which is specifically related with SMS1 but not SMS2 deficiency. To our surprise, hepatocyte total SMS deficiency has marginal effect on hepatocyte ceramide, diacylglyceride, and phosphatidylcholine levels. Importantly, total SMS deficiency decreases plasma triglyceride but not apoB levels and reduces larger VLDL concentration. The reduction of triglyceride levels also was observed when the animals were on a high fat diet. Our results show that hepatocyte total SMS blocking can reduce VLDL-triglyceride production and plasma triglyceride levels. This phenomenon could be related with a reduction of atherogenicity.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Yeun-Po Chiang
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Mulin He
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Ke Zhang
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Jiao Zheng
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Weihua Wu
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Jiajia Cai
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Yong Chen
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | - Guangzhi Chen
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America
| | | | | | - Tilla S Worgall
- Department of Medicine, Columbia University, United States of America
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, United States of America; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, United States of America.
| |
Collapse
|
6
|
Jiang XC, Yu Y. The Role of Phospholipid Transfer Protein in the Development of Atherosclerosis. Curr Atheroscler Rep 2021; 23:9. [PMID: 33496859 DOI: 10.1007/s11883-021-00907-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Phospholipid transfer protein (PLTP), a member of lipid transfer protein family, is an important protein involved in lipid metabolism in the circulation. This article reviews recent PLTP research progresses, involving lipoprotein metabolism and atherogenesis. RECENT FINDINGS PLTP activity influences atherogenic and anti-atherogenic lipoprotein levels. Human serum PLTP activity is a risk factor for human cardiovascular disease and is an independent predictor of all-cause mortality. PLTP deficiency reduces VLDL and LDL levels and attenuates atherosclerosis in mouse models, while PLTP overexpression exerts an opposite effect. Both PLTP deficiency and overexpression result in reduction of HDL which has different size, inflammatory index, and lipid composition. Moreover, although both PLTP deficiency and overexpression reduce cholesterol efflux capacity, but this effect has no impact in macrophage reverse cholesterol transport in mice. Furthermore, PLTP activity is related with metabolic syndrome, thrombosis, and inflammation. PLTP could be target for the treatment of dyslipidemia and atherosclerosis, although some potential off-target effects should be noted.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, USA.
| | - Yang Yu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| |
Collapse
|
7
|
He X, Qi Z, Hou H, Gao J, Zhang XX. Effects of chronic cadmium exposure at food limitation-relevant levels on energy metabolism in mice. JOURNAL OF HAZARDOUS MATERIALS 2020; 388:121791. [PMID: 31818666 DOI: 10.1016/j.jhazmat.2019.121791] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 11/25/2019] [Accepted: 11/29/2019] [Indexed: 06/10/2023]
Abstract
Cadmium (Cd) exposure has been implicated in the perturbation of energy metabolism and the development of cardiometabolic disease, but disease predisposition from chronic low-dose Cd exposure remains unclear. This study employed a mouse model to investigate the toxic effects of chronic Cd exposure at food limitation-relevant levels on energy metabolism and the associated liver and gut microbiome functions. Results showed that the Cd exposure induced the perturbation of energy metabolism in mice, evidenced by the alteration of various metabolites associated with the phosphorogen (adenosine triphosphate-creatine phosphate) system, tricarboxylic acid cycle, and lipid metabolism, as well as the increase of the cardiometabolic risk factor, triglyceride. Moreover, both liver and gut microbiome underwent marked structural/histological and functional alterations, prone to the onset of cardiometabolic disease following the Cd exposure. Certain hepatic transcription factors and gut microbes, specifically PPARα, SREBP1c, HNF4A and the Clostridiales_vadinBB60_group, were identified to be highly correlated with altered urinary metabolites, revealing potential toxicological interactions between the liver and gut microbiome, and energy metabolism. Our findings provide new insights into the progression of metabolic diseases induced by Cd exposure. We also propose a stricter Cd limitation in future food safety standards.
Collapse
Affiliation(s)
- Xiwei He
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Zhaodong Qi
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hui Hou
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Jie Gao
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Xu-Xiang Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
8
|
Impact of Phospholipid Transfer Protein in Lipid Metabolism and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:1-13. [PMID: 32705590 DOI: 10.1007/978-981-15-6082-8_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PLTP plays an important role in lipoprotein metabolism and cardiovascular disease development in humans; however, the mechanisms are still not completely understood. In mouse models, PLTP deficiency reduces cardiovascular disease, while its overexpression induces it. Therefore, we used mouse models to investigate the involved mechanisms. In this chapter, the recent main progresses in the field of PLTP research are summarized, and our focus is on the relationship between PLTP and lipoprotein metabolism, as well as PLTP and cardiovascular diseases.
Collapse
|
9
|
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
|
10
|
Fukui H. Role of Gut Dysbiosis in Liver Diseases: What Have We Learned So Far? Diseases 2019; 7:diseases7040058. [PMID: 31726747 PMCID: PMC6956030 DOI: 10.3390/diseases7040058] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence supports that gut dysbiosis may relate to various liver diseases. Alcoholics with high intestinal permeability had a decrease in the abundance of Ruminnococcus. Intestinal dysmotility, increased gastric pH, and altered immune responses in addition to environmental and genetic factors are likely to cause alcohol-associated gut microbial changes. Alcohol-induced dysbiosis may be associated with gut barrier dysfunction, as microbiota and their products modulate barrier function by affecting epithelial pro-inflammatory responses and mucosal repair functions. High levels of plasma endotoxin are detected in alcoholics, in moderate fatty liver to advanced cirrhosis. Decreased abundance of Faecalibacterium prausnitzii, an anti-inflammatory commensal, stimulating IL-10 secretion and inhibiting IL-12 and interferon-γ expression. Proteobacteria, Enterobacteriaceae, and Escherichia were reported to be increased in NAFLD (nonalcoholic fatty liver disease) patients. Increased abundance of fecal Escherichia to elevated blood alcohol levels in these patients and gut microbiota enriched in alcohol-producing bacteria produce more alcohol (alcohol hypothesis). Some undetermined pathological sequences related to gut dysbiosis may facilitate energy-producing and proinflammatory conditions for the progression of NAFLD. A shortage of autochthonous non-pathogenic bacteria and an overgrowth of potentially pathogenic bacteria are common findings in cirrhotic patients. The ratio of the amounts of beneficial autochthonous taxa (Lachnospiraceae + Ruminococaceae + Veillonellaceae + Clostridiales Incertae Sedis XIV) to those of potentially pathogenic taxa (Enterobacteriaceae + Bacteroidaceae) was low in those with early death and organ failure. Cirrhotic patients with decreased microbial diversity before liver transplantation were more likely to develop post-transplant infections and cognitive impairment related to residual dysbiosis. Patients with PSC had marked reduction of bacterial diversity. Enterococcus and Lactobacillus were increased in PSC patients (without liver cirrhosis.) Treatment-naive PBC patients were associated with altered composition and function of gut microbiota, as well as a lower level of diversity. As serum anti-gp210 antibody has been considered as an index of disease progression, relatively lower species richness and lower abundance of Faecalibacterium spp. in gp210-positive patients are interesting. The dysbiosis-induced altered bacterial metabolites such as a hepatocarcinogenesis promotor DCA, together with a leaky gut and bacterial translocation. Gut protective Akkermansia and butyrate-producing genera were decreased, while genera producing-lipopolysaccharide were increased in early hepatocellular carcinoma (HCC) patients.
Collapse
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8522, Japan
| |
Collapse
|
11
|
Role of Gut Microbiota in Hepatocarcinogenesis. Microorganisms 2019; 7:microorganisms7050121. [PMID: 31060311 PMCID: PMC6560397 DOI: 10.3390/microorganisms7050121] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/23/2019] [Accepted: 05/03/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC), one of the leading causes of death worldwide, has a causal nexus with liver injury, inflammation, and regeneration that accumulates over decades. Observations from recent studies have accounted for the involvement of the gut–liver axis in the pathophysiological mechanism responsible for HCC. The human intestine nurtures a diversified colony of microorganisms residing in the host ecosystem. The intestinal barrier is critical for conserving the normal physiology of the gut microbiome. Therefore, a rupture of this barrier or dysbiosis can cause the intestinal microbiome to serve as the main source of portal-vein endotoxins, such as lipopolysaccharide, in the progression of hepatic diseases. Indeed, increased bacterial translocation is a key sign of HCC. Considering the limited number of clinical studies on HCC with respect to the microbiome, we focus on clinical as well as animal studies involving the gut microbiota, with the current understandings of the mechanism by which the intestinal dysbiosis promotes hepatocarcinogenesis. Future research might offer mechanistic insights into the specific phyla targeting the leaky gut, as well as microbial dysbiosis, and their metabolites, which represent key pathways that drive HCC-promoting microbiome-mediated liver inflammation and fibrosis, thereby restoring the gut barrier function.
Collapse
|
12
|
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
|
13
|
Yu Y, Lei X, Jiang H, Li Z, Creemers JWM, Zhang M, Qin S, Jin W, Jiang X. Prodomain of Furin Promotes Phospholipid Transfer Protein Proteasomal Degradation in Hepatocytes. J Am Heart Assoc 2018; 7:e008526. [PMID: 29680823 PMCID: PMC6015287 DOI: 10.1161/jaha.118.008526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/22/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Phospholipid transfer protein (PLTP) is one of the major modulators of lipoprotein metabolism and atherosclerosis development; however, little is known about the regulation of PLTP. The effect of hepatic prodomain of furin (profurin) expression on PLTP processing and function is investigated. METHODS AND RESULTS We used adenovirus expressing profurin in mouse liver to evaluate PLTP activity, mass, and plasma lipid levels. We coexpressed PLTP and profurin in human hepatoma cell line cells and studied their interaction. We found profurin expression significantly reduced plasma lipids, plasma PLTP activity, and mass in all tested mouse models, compared with controls. Moreover, the expression of profurin dramatically reduced liver PLTP activity and protein level. We further explored the mechanism using in vivo and ex vivo approaches. We found that profurin can interact with intracellular PLTP and promote its ubiquitination and proteasomal degradation, resulting in less PLTP secretion from the hepatocytes. Furin does not cleave PLTP; instead, it forms a complex with PLTP, likely through its prodomain. CONCLUSIONS Our study reveals that hepatic PLTP protein is targeted for proteasomal degradation by profurin expression, which could be a novel posttranslational mechanism underlying PLTP regulation.
Collapse
Affiliation(s)
- Yang Yu
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Xia Lei
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Hui Jiang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Zhiqiang Li
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - John W. M. Creemers
- Laboratory of Biochemical NeuroendocrinologyDepartment of Human GeneticsHerestraat 49 bus 6023000 LeuvenBelgium
| | - Ming Zhang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Weijun Jin
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Xian‐Cheng Jiang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
- Molecular and Cellular Cardiology ProgramVeterans Affair New York Harbor Healthcare SystemBrooklynNY
| |
Collapse
|
14
|
Jiang XC. Phospholipid transfer protein: its impact on lipoprotein homeostasis and atherosclerosis. J Lipid Res 2018; 59:764-771. [PMID: 29438986 DOI: 10.1194/jlr.r082503] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/10/2018] [Indexed: 12/25/2022] Open
Abstract
Phospholipid transfer protein (PLTP) is one of the major modulators of lipoprotein metabolism and atherosclerosis development in humans; however, we still do not quite understand the mechanisms. In mouse models, PLTP overexpression induces atherosclerosis, while its deficiency reduces it. Thus, mouse models were used to explore the mechanisms. In this review, I summarize the major progress made in the PLTP research field and emphasize its impact on lipoprotein metabolism and atherosclerosis, as well as its regulation.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, Downstate Medical Center, State University of New York, Brooklyn, NY
| |
Collapse
|
15
|
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of worldwide cancer mortality. HCC almost exclusively develops in patients with chronic liver disease, driven by a vicious cycle of liver injury, inflammation and regeneration that typically spans decades. Increasing evidence points towards a key role of the bacterial microbiome in promoting the progression of liver disease and the development of HCC. Here, we will review mechanisms by which the gut microbiota promotes hepatocarcinogenesis, focusing on the leaky gut, bacterial dysbiosis, microbe-associated molecular patterns and bacterial metabolites as key pathways that drive cancer-promoting liver inflammation, fibrosis and genotoxicity. On the basis of accumulating evidence from preclinical studies, we propose the intestinal-microbiota-liver axis as a promising target for the simultaneous prevention of chronic liver disease progression and HCC development in patients with advanced liver disease. We will review in detail therapeutic modalities and discuss clinical settings in which targeting the gut-microbiota-liver axis for the prevention of disease progression and HCC development seems promising.
Collapse
Affiliation(s)
- Le-Xing Yu
- Department of Medicine, Columbia University, 1130 St. Nicholas Avenue, Room 926, New York, New York 10032, USA
| | - Robert F Schwabe
- Department of Medicine, Columbia University, 1130 St. Nicholas Avenue, Room 926, New York, New York 10032, USA
- Institute of Human Nutrition, 1130 St. Nicholas Avenue, Room 926, New York, New York 10032, USA
| |
Collapse
|
16
|
Yang Y, Zhang H, Yan B, Zhang T, Gao Y, Shi Y, Le G. Health Effects of Dietary Oxidized Tyrosine and Dityrosine Administration in Mice with Nutrimetabolomic Strategies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:6957-6971. [PMID: 28742334 DOI: 10.1021/acs.jafc.7b02003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This study aims to investigate the health effects of long-term dietary oxidized tyrosine (O-Tyr) and its main product (dityrosine) administration on mice metabolism. Mice received daily intragastric administration of either O-Tyr (320 μg/kg body weight), dityrosine (Dityr, 320 μg/kg body weight), or saline for consecutive 6 weeks. Urine and plasma samples were analyzed by NMR-based metabolomics strategies. Body weight, clinical chemistry, oxidative damage indexes, and histopathological data were obtained as complementary information. O-Tyr and Dityr exposure changed many systemic metabolic processes, including reduced choline bioavailability, led to fat accumulation in liver, induced hepatic injury, and renal dysfunction, resulted in changes in gut microbiota functions, elevated risk factor for cardiovascular disease, altered amino acid metabolism, induced oxidative stress responses, and inhibited energy metabolism. These findings implied that it is absolutely essential to reduce the generation of oxidation protein products in food system through improving modern food processing methods.
Collapse
Affiliation(s)
- Yuhui Yang
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Hui Zhang
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Biao Yan
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Tianyu Zhang
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Ying Gao
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Yonghui Shi
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
- The State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| | - Guowei Le
- The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
- The State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University , Wuxi, Jiangsu 214122, China
| |
Collapse
|
17
|
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
|
18
|
Gut Microbiota and Lifestyle Interventions in NAFLD. Int J Mol Sci 2016; 17:447. [PMID: 27023533 PMCID: PMC4848903 DOI: 10.3390/ijms17040447] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Abstract
The human digestive system harbors a diverse and complex community of microorganisms that work in a symbiotic fashion with the host, contributing to metabolism, immune response and intestinal architecture. However, disruption of a stable and diverse community, termed "dysbiosis", has been shown to have a profound impact upon health and disease. Emerging data demonstrate dysbiosis of the gut microbiota to be linked with non-alcoholic fatty liver disease (NAFLD). Although the exact mechanism(s) remain unknown, inflammation, damage to the intestinal membrane, and translocation of bacteria have all been suggested. Lifestyle intervention is undoubtedly effective at improving NAFLD, however, not all patients respond to these in the same manner. Furthermore, studies investigating the effects of lifestyle interventions on the gut microbiota in NAFLD patients are lacking. A deeper understanding of how different aspects of lifestyle (diet/nutrition/exercise) affect the host-microbiome interaction may allow for a more tailored approach to lifestyle intervention. With gut microbiota representing a key element of personalized medicine and nutrition, we review the effects of lifestyle interventions (diet and physical activity/exercise) on gut microbiota and how this impacts upon NAFLD prognosis.
Collapse
|
19
|
Oliveira CP, de Lima Sanches P, de Abreu-Silva EO, Marcadenti A. Nutrition and Physical Activity in Nonalcoholic Fatty Liver Disease. J Diabetes Res 2016; 2016:4597246. [PMID: 26770987 PMCID: PMC4685119 DOI: 10.1155/2016/4597246] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 07/26/2015] [Accepted: 07/29/2015] [Indexed: 12/26/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide and it is associated with other medical conditions such as diabetes mellitus, metabolic syndrome, and obesity. The mechanisms of the underlying disease development and progression are not completely established and there is no consensus concerning the pharmacological treatment. In the gold standard treatment for NAFLD weight loss, dietary therapy, and physical activity are included. However, little scientific evidence is available on diet and/or physical activity and NAFLD specifically. Many dietary approaches such as Mediterranean and DASH diet are used for treatment of other cardiometabolic risk factors such as insulin resistance and type-2 diabetes mellitus (T2DM), but on the basis of its components their role in NAFLD has been discussed. In this review, the implications of current dietary and exercise approaches, including Brazilian and other guidelines, are discussed, with a focus on determining the optimal nonpharmacological treatment to prescribe for NAFLD.
Collapse
Affiliation(s)
- Claudia P. Oliveira
- Department of Gastroenterology, School of Medicine, University of Sao Paulo (USP), 255 Dr. Enéas de Carvalho Aguiar Avenue, Cerqueira César, 05403-900 Sao Paulo, SP, Brazil
| | - Priscila de Lima Sanches
- Postgraduate Program in Nutrition, Federal University of Sao Paulo (UNIFESP), 630 Marselhesa Street, Vila Clementino, 04020-060 Sao Paulo, SP, Brazil
| | - Erlon Oliveira de Abreu-Silva
- Division of Interventional Cardiology and Postgraduate Program in Cardiology, Federal University of Sao Paulo (UNIFESP), 715 Napoleao de Barros Street, Vila Clementino, 04024-002 Sao Paulo, SP, Brazil
| | - Aline Marcadenti
- Department of Nutrition, Federal University of Health Sciences of Porto Alegre (UFCSPA), 245 Sarmento Leite Street, Centro Histórico, 90050-170 Porto Alegre, RS, Brazil
- Postgraduate Program in Health Sciences: Cardiology, Institute of Cardiology of Rio Grande do Sul (IC/FUC), 395 Princesa Isabel Avenue, Santana, 90040-371 Porto Alegre, RS, Brazil
- *Aline Marcadenti:
| |
Collapse
|
20
|
Gut Microbiota and Host Reaction in Liver Diseases. Microorganisms 2015; 3:759-91. [PMID: 27682116 PMCID: PMC5023261 DOI: 10.3390/microorganisms3040759] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/08/2015] [Accepted: 10/21/2015] [Indexed: 02/07/2023] Open
Abstract
Although alcohol feeding produces evident intestinal microbial changes in animals, only some alcoholics show evident intestinal dysbiosis, a decrease in Bacteroidetes and an increase in Proteobacteria. Gut dysbiosis is related to intestinal hyperpermeability and endotoxemia in alcoholic patients. Alcoholics further exhibit reduced numbers of the beneficial Lactobacillus and Bifidobacterium. Large amounts of endotoxins translocated from the gut strongly activate Toll-like receptor 4 in the liver and play an important role in the progression of alcoholic liver disease (ALD), especially in severe alcoholic liver injury. Gut microbiota and bacterial endotoxins are further involved in some of the mechanisms of nonalcoholic fatty liver disease (NAFLD) and its progression to nonalcoholic steatohepatitis (NASH). There is experimental evidence that a high-fat diet causes characteristic dysbiosis of NAFLD, with a decrease in Bacteroidetes and increases in Firmicutes and Proteobacteria, and gut dysbiosis itself can induce hepatic steatosis and metabolic syndrome. Clinical data support the above dysbiosis, but the details are variable. Intestinal dysbiosis and endotoxemia greatly affect the cirrhotics in relation to major complications and prognosis. Metagenomic approaches to dysbiosis may be promising for the analysis of deranged host metabolism in NASH and cirrhosis. Management of dysbiosis may become a cornerstone for the future treatment of liver diseases.
Collapse
|
21
|
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
|
22
|
Abdul-Hai A, Abdallah A, Malnick SDH. Influence of gut bacteria on development and progression of non-alcoholic fatty liver disease. World J Hepatol 2015; 7:1679-1684. [PMID: 26140087 PMCID: PMC4483549 DOI: 10.4254/wjh.v7.i12.1679] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/20/2015] [Accepted: 04/20/2015] [Indexed: 02/06/2023] Open
Abstract
The intestine of the human contains a dynamic population of microbes that have a symbiotic relationship with the host. In addition, there is an effect of the intestinal microbiota on metabolism and digestion. Non-alcoholic fatty liver disease (NAFLD) is a common cause worldwide of hepatic pathology and is thought to be the hepatic manifestation of the metabolic syndrome. In this review we examine the effect of the human microbiome on the components and pathogenesis of the metabolic syndrome. We are now on the threshold of therapeutic interventions on the human microbiome in order to effect human disease including NAFLD.
Collapse
|
23
|
Abstract
The leaky gut hypothesis links translocating microbial products with the onset and progression of liver disease, and for a long time was considered one of its major contributors. However, a more detailed picture of the intestinal microbiota contributing to liver disease started to evolve. The gut is colonized by trillions of microbes that aid in digestion, modulate immune response, and generate a variety of products that result from microbial metabolic activities. These products together with host-bacteria interactions influence both normal physiology and disease susceptibility. A disruption of the symbiosis between microbiota and host is known as dysbiosis and can have profound effects on health. Qualitative changes such as increased proportions of harmful bacteria and reduced levels of beneficial bacteria, and also quantitative changes in the total amount of bacteria (overgrowth) have been associated with liver disease. Understanding the link between the pathophysiology of liver diseases and compositional and functional changes of the microbiota will help in the design of innovative therapies. In this review, we focus on factors resulting in dysbiosis, and discuss how dysbiosis can disrupt intestinal homeostasis and contribute to liver disease.
Collapse
Key Words
- dysbiosis
- leaky gut
- alcoholic liver disease
- nash
- nafld
- cirrhosis
- microbiome
- pamps
- ald, alcoholic liver disease
- amp, antimicrobial peptides and proteins
- fiaf, fasting-induced adipocyte factor
- hfd, high-fat diet
- ibd, inflammatory bowel disease
- il, interleukin
- lcfa, long-chain fatty acid
- lps, lipopolysaccharide
- nafld, nonalcoholic fatty liver disease
- nash, nonalcoholic steatohepatitis
- nlrp, nucleotide-binding domain and leucine rich repeat-containing protein
- nod2, nucleotide-binding oligomerization domain 2
- pamps, pathogen-associated molecular patterns
- reg3, regenerating islet-derived 3
- tlr, toll-like receptor
- tnf, tumor necrosis factor
- tnfr, tumor necrosis factor receptor
Collapse
Affiliation(s)
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, and Department of Medicine, VA San Diego Healthcare System, San Diego, California
| |
Collapse
|
24
|
Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014; 146:1513-24. [PMID: 24440671 PMCID: PMC3996054 DOI: 10.1053/j.gastro.2014.01.020] [Citation(s) in RCA: 728] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/29/2013] [Accepted: 01/07/2014] [Indexed: 02/08/2023]
Abstract
The human intestine harbors a diverse community of microbes that promote metabolism and digestion in their symbiotic relationship with the host. Disturbance of its homeostasis can result in disease. We review factors that disrupt intestinal homeostasis and contribute to nonalcoholic fatty liver disease, steatohepatitis, alcoholic liver disease, and cirrhosis. Liver disease has long been associated with qualitative and quantitative (overgrowth) dysbiotic changes in the intestinal microbiota. Extrinsic factors, such as the Western diet and alcohol, contribute to these changes. Dysbiosis results in intestinal inflammation, a breakdown of the intestinal barrier, and translocation of microbial products in animal models. However, the contribution of the intestinal microbiome to liver disease goes beyond simple translocation of bacterial products that promote hepatic injury and inflammation. Microbial metabolites produced in a dysbiotic intestinal environment and host factors are equally important in the pathogenesis of liver disease. We review how the combination of liver insult and disruptions in intestinal homeostasis contribute to liver disease.
Collapse
Affiliation(s)
- Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California.
| | | |
Collapse
|
25
|
Yazdanyar A, Quan W, Jin W, Jiang XC. Liver-specific phospholipid transfer protein deficiency reduces high-density lipoprotein and non-high-density lipoprotein production in mice. Arterioscler Thromb Vasc Biol 2013; 33:2058-64. [PMID: 23846500 DOI: 10.1161/atvbaha.113.301628] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The liver is one of the critical organs for lipoprotein metabolism and a major source for phospholipid transfer protein (PLTP) expression. The effect of liver-specific PLTP deficiency on plasma lipoprotein production and metabolism in mice was investigated. APPROACH AND RESULTS We created a liver-specific PLTP-deficient mouse model. We measured plasma high-density lipoprotein (HDL) and apolipoprotein B (apoB)-containing lipoprotein (or non-HDL) levels and their production rates. We found that hepatic ablation of PLTP leads to a significant decrease in plasma PLTP activity, HDL lipids, non-HDL lipids, apoAI, and apoB levels. In addition, nuclear magnetic resonance examination of lipoproteins showed that the deficiency decreases HDL and apoB-containing lipoprotein particle numbers, as well as very low-density lipoprotein particle size, which was confirmed by electron microscopy. Moreover, HDL particles from the deficient mice are lipid-poor ones. To unravel the mechanism, we evaluated the apoB and triglyceride production rates. We found that hepatic PLTP deficiency significantly decreases apoB and triglyceride secretion rates. To investigate the role of liver PLTP on HDL production, we set up primary hepatocyte culture studies and found that the PLTP-deficient hepatocytes produce less nascent HDL. Furthermore, we found that exogenous PLTP promotes nascent HDL production through an ATP-binding cassette A 1-mediated pathway. CONCLUSIONS Liver-specific PLTP deficiency significantly reduces plasma HDL and apoB-containing lipoprotein levels. Reduction of production rates of both particles is one of the mechanisms.
Collapse
Affiliation(s)
- Amirfarbod Yazdanyar
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | | | | | | |
Collapse
|
26
|
Endoplasmic reticulum-localized hepatic lipase decreases triacylglycerol storage and VLDL secretion. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1113-23. [DOI: 10.1016/j.bbalip.2013.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 01/09/2013] [Accepted: 01/23/2013] [Indexed: 01/07/2023]
|
27
|
Proteomics and gene expression analyses of squalene-supplemented mice identify microsomal thioredoxin domain-containing protein 5 changes associated with hepatic steatosis. J Proteomics 2012; 77:27-39. [DOI: 10.1016/j.jprot.2012.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 06/28/2012] [Accepted: 07/01/2012] [Indexed: 11/18/2022]
|
28
|
Jiang XC, Jin W, Hussain MM. The impact of phospholipid transfer protein (PLTP) on lipoprotein metabolism. Nutr Metab (Lond) 2012; 9:75. [PMID: 22897926 PMCID: PMC3495888 DOI: 10.1186/1743-7075-9-75] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/30/2012] [Indexed: 02/05/2023] Open
Abstract
It has been reported that phospholipid transfer protein (PLTP) is an independent risk factor for human coronary artery disease. In mouse models, it has been demonstrated that PLTP overexpression induces atherosclerosis, while its deficiency reduces it. PLTP is considered a promising target for pharmacological intervention to treat atherosclerosis. However, we must still answer a number of questions before its pharmaceutical potential can be fully explored. In this review, we summarized the recent progresses made in the PLTP research field and focused on its effect on apoB-containing- triglyceride-rich particle and HDL metabolism.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, Downstate Medical Center, State University of New York, 450 Clarkson Ave,, Box 5, Brooklyn, NY, 11203, USA.
| | | | | |
Collapse
|
29
|
Yazdanyar A, Jiang XC. Liver phospholipid transfer protein (PLTP) expression with a PLTP-null background promotes very low-density lipoprotein production in mice. Hepatology 2012; 56:576-84. [PMID: 22367708 PMCID: PMC3409695 DOI: 10.1002/hep.25648] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 01/31/2012] [Indexed: 12/07/2022]
Abstract
UNLABELLED It is known that plasma phospholipid transfer protein (PLTP) activity influences lipoprotein metabolism. The liver is one of the major sites of lipoprotein production and degradation, as well as of PLTP expression. To address the impact of liver-expressed PLTP on lipoprotein metabolism, we created a mouse model that expresses PLTP in the liver acutely and specifically, with a PLTP-null background. This approach in mouse model preparations can also be used universally for evaluating the function of many other genes in the liver. We found that liver PLTP expression dramatically increases plasma levels of non-high-density lipoprotein (HDL) cholesterol (2.7-fold, P < 0.0001), non-HDL phospholipid (2.5-fold, P < 0.001), and triglyceride (51%, P < 0.01), but has no significant influence on plasma HDL lipids compared with controls. Plasma apolipoprotein (apo)B levels were also significantly increased in PLTP-expressing mice (2.2-fold, P < 0.001), but those of apoA-I were not. To explore the mechanism involved, we examined the lipidation and secretion of nascent very low-density lipoprotein (VLDL), finding that liver PLTP expression significantly increases VLDL lipidation in hepatocyte microsomal lumina, and also VLDL secretion into the plasma. CONCLUSION It is possible to prepare a mouse model that expresses the gene of interest only in the liver, but not in other tissues. Our results suggest, for the first time, that the major function of liver PLTP is to drive VLDL production and makes a small contribution to plasma PLTP activity.
Collapse
Affiliation(s)
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center
- To whom correspondence may be addressed at Downstate Medical Center, 450 Clarkson Ave. Box 5 Brooklyn, NY 11203, tel. (718) 270-6701, Fax (718) 270-3732,
| |
Collapse
|
30
|
Rosenson RS, Brewer HB, Davidson WS, Fayad ZA, Fuster V, Goldstein J, Hellerstein M, Jiang XC, Phillips MC, Rader DJ, Remaley AT, Rothblat GH, Tall AR, Yvan-Charvet L. Cholesterol efflux and atheroprotection: advancing the concept of reverse cholesterol transport. Circulation 2012; 125:1905-19. [PMID: 22508840 PMCID: PMC4159082 DOI: 10.1161/circulationaha.111.066589] [Citation(s) in RCA: 724] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Robert S Rosenson
- Mount Sinai Heart, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Li Z, Ding T, Pan X, Li Y, Li R, Sanders PE, Kuo MS, Hussain MM, Cao G, Jiang XC. Lysophosphatidylcholine acyltransferase 3 knockdown-mediated liver lysophosphatidylcholine accumulation promotes very low density lipoprotein production by enhancing microsomal triglyceride transfer protein expression. J Biol Chem 2012; 287:20122-31. [PMID: 22511767 DOI: 10.1074/jbc.m111.334664] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
After de novo biosynthesis phospholipids undergo extensive remodeling by the Lands' cycle. Enzymes involved in phospholipid biosynthesis have been studied extensively but not those involved in reacylation of lysophosphopholipids. One key enzyme in the Lands' cycle is fatty acyl-CoA:lysophosphatidylcholine acyltransferase (LPCAT), which utilizes lysophosphatidylcholine (LysoPC) and fatty acyl-CoA to produce various phosphatidylcholine (PC) species. Four isoforms of LPCAT have been identified. In this study we found that LPCAT3 is the major hepatic isoform, and its knockdown significantly reduces hepatic LPCAT activity. Moreover, we report that hepatic LPCAT3 knockdown increases certain species of LysoPCs and decreases certain species of PC. A surprising observation was that LPCAT3 knockdown significantly reduces hepatic triglycerides. Despite this, these mice had higher plasma triglyceride and apoB levels. Lipoprotein production studies indicated that reductions in LPCAT3 enhanced assembly and secretion of triglyceride-rich apoB-containing lipoproteins. Furthermore, these mice had higher microsomal triglyceride transfer protein (MTP) mRNA and protein levels. Mechanistic studies in hepatoma cells revealed that LysoPC enhances secretion of apoB but not apoA-I in a concentration-dependent manner. Moreover, LysoPC increased MTP mRNA, protein, and activity. In short, these results indicate that hepatic LPCAT3 modulates VLDL production by regulating LysoPC levels and MTP expression.
Collapse
Affiliation(s)
- Zhiqiang Li
- Molecular and Cellular Cardiology Program, Veterans Affairs New York Harbor Healthcare System, Brooklyn, New York 11209, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Plasma PLTP (phospholipid-transfer protein): an emerging role in 'reverse lipopolysaccharide transport' and innate immunity. Biochem Soc Trans 2011; 39:984-8. [PMID: 21787334 DOI: 10.1042/bst0390984] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Plasma PLTP (phospholipid-transfer protein) is a member of the lipid transfer/LBP [LPS (lipopolysaccharide)-binding protein] family, which constitutes a superfamily of genes together with the short and long PLUNC (palate, lung and nasal epithelium clone) proteins. Although PLTP was studied initially for its involvement in the metabolism of HDL (high-density lipoproteins) and reverse cholesterol transport (i.e. the metabolic pathway through which cholesterol excess can be transported from peripheral tissues back to the liver for excretion in the bile), it displays a number of additional biological properties. In particular, PLTP can modulate the lipoprotein association and metabolism of LPS that are major components of Gram-negative bacteria. The delayed association of LPS with lipoproteins in PLTP-deficient mice results in a prolonged residence time, in a higher toxicity of LPS aggregates and in a significant increase in LPS-induced mortality as compared with wild-type mice. It suggests that PLTP may play a pivotal role in inflammation and innate immunity through its ability to accelerate the 'reverse LPS transport' pathway.
Collapse
|
33
|
Yazdanyar A, Yeang C, Jiang XC. Role of phospholipid transfer protein in high-density lipoprotein- mediated reverse cholesterol transport. Curr Atheroscler Rep 2011; 13:242-8. [PMID: 21365262 PMCID: PMC3085729 DOI: 10.1007/s11883-011-0172-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Reverse cholesterol transport (RCT) describes the process whereby cholesterol in peripheral tissues is transported to the liver where it is ultimately excreted in the form of bile. Given the atherogenic role of cholesterol accumulation within the vessel intima, removal of cholesterol through RCT is considered an anti-atherogenic process. The major constituents of RCT include cell membrane– bound lipid transporters, plasma lipid acceptors, plasma proteins and enzymes, and lipid receptors of liver cell membrane. One major cholesterol acceptor in RCT is high-density lipoprotein (HDL). Both the characteristics and level of HDL are critical determinants for RCT. It is known that phospholipid transfer protein (PLTP) impacts both HDL cholesterol level and biological quality of the HDL molecule. Recent data suggest that PLTP has a site-specific variation in its function. Moreover, the RCT pathway also has multiple steps both in the peripheral tissues and circulation. Therefore, PLTP may influence the RCT pathway at multiple levels. In this review, we focus on the potential role of PLTP in RCT through its impact on HDL homeostasis. The relationship between PLTP and RCT is expected to be an important area in finding novel therapies for atherosclerosis.
Collapse
Affiliation(s)
- Amirfarbod Yazdanyar
- Department of cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave. Box 5, Brooklyn, NY 11203, USA
| | | | | |
Collapse
|
34
|
Cole LK, Vance JE, Vance DE. Phosphatidylcholine biosynthesis and lipoprotein metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:754-61. [PMID: 21979151 DOI: 10.1016/j.bbalip.2011.09.009] [Citation(s) in RCA: 263] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 09/15/2011] [Accepted: 09/19/2011] [Indexed: 12/28/2022]
Abstract
Phosphatidylcholine (PC) is the major phospholipid component of all plasma lipoprotein classes. PC is the only phospholipid which is currently known to be required for lipoprotein assembly and secretion. Impaired hepatic PC biosynthesis significantly reduces the levels of circulating very low density lipoproteins (VLDLs) and high density lipoproteins (HDLs). The reduction in plasma VLDLs is due in part to impaired hepatic secretion of VLDLs. Less PC within the hepatic secretory pathway results in nascent VLDL particles with reduced levels of PC. These particles are recognized as being defective and are degraded within the secretory system by an incompletely defined process that occurs in a post-endoplasmic reticulum compartment, consistent with degradation directed by the low-density lipoprotein receptor and/or autophagy. Moreover, VLDL particles are taken up more readily from the circulation when the PC content of the VLDLs is reduced, likely due to a preference of cell surface receptors and/or enzymes for lipoproteins that contain less PC. Impaired PC biosynthesis also reduces plasma HDLs by inhibiting hepatic HDL formation and by increasing HDL uptake from the circulation. These effects are mediated by elevated expression of ATP-binding cassette transporter A1 and hepatic scavenger receptor class B type 1, respectively. Hepatic PC availability has recently been linked to the progression of liver and heart disease. These findings demonstrate that hepatic PC biosynthesis can regulate the amount of circulating lipoproteins and suggest that hepatic PC biosynthesis may represent an important pharmaceutical target. This article is part of a Special Issue entitled Triglyceride Metabolism and Disease.
Collapse
Affiliation(s)
- Laura K Cole
- Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
35
|
Gambino R, Musso G, Cassader M. Redox balance in the pathogenesis of nonalcoholic fatty liver disease: mechanisms and therapeutic opportunities. Antioxid Redox Signal 2011; 15:1325-65. [PMID: 20969475 DOI: 10.1089/ars.2009.3058] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is currently the most common liver disease in the world. It encompasses a histological spectrum, ranging from simple, nonprogressive steatosis to nonalcoholic steatohepatitis (NASH), which may progress to cirrhosis and hepatocellular carcinoma. While liver-related complications are confined to NASH, emerging evidence suggests both simple steatosis and NASH predispose to type 2 diabetes and cardiovascular disease. The pathogenesis of NAFLD is currently unknown, but accumulating data suggest that oxidative stress and altered redox balance play a crucial role in the pathogenesis of steatosis, steatohepatitis, and fibrosis. We will examine intracellular mechanisms, including mitochondrial dysfunction and impaired oxidative free fatty acid metabolism, leading to reactive oxygen species generation; additionally, the potential pathogenetic role of extracellular sources of reactive oxygen species in NAFLD, including increased myeloperoxidase activity and oxidized low density lipoprotein accumulation, will be reviewed. We will discuss how these mechanisms converge to determine the whole pathophysiological spectrum of NAFLD, including hepatocyte triglyceride accumulation, hepatocyte apoptosis, hepatic inflammation, hepatic stellate cell activation, and fibrogenesis. Finally, available animal and human data on treatment opportunities with older and newer antioxidant will be presented.
Collapse
Affiliation(s)
- Roberto Gambino
- Department of Internal Medicine, University of Turin, Turin, Italy
| | | | | |
Collapse
|
36
|
Abstract
It has been shown that inhibition of de novo sphingolipid synthesis increases insulin sensitivity. For further exploration of the mechanism involved, we utilized two models: heterozygous serine palmitoyltransferase (SPT) subunit 2 (Sptlc2) gene knockout mice and sphingomyelin synthase 2 (Sms2) gene knockout mice. SPT is the key enzyme in sphingolipid biosynthesis, and Sptlc2 is one of its subunits. Homozygous Sptlc2-deficient mice are embryonic lethal. However, heterozygous Sptlc2-deficient mice that were viable and without major developmental defects demonstrated decreased ceramide and sphingomyelin levels in the cell plasma membranes, as well as heightened sensitivity to insulin. Moreover, these mutant mice were protected from high-fat diet-induced obesity and insulin resistance. SMS is the last enzyme for sphingomyelin biosynthesis, and SMS2 is one of its isoforms. Sms2 deficiency increased cell membrane ceramide but decreased SM levels. Sms2 deficiency also increased insulin sensitivity and ameliorated high-fat diet-induced obesity. We have concluded that Sptlc2 heterozygous deficiency- or Sms2 deficiency-mediated reduction of SM in the plasma membranes leads to an improvement in tissue and whole-body insulin sensitivity.
Collapse
|
37
|
Albers JJ, Vuletic S, Cheung MC. Role of plasma phospholipid transfer protein in lipid and lipoprotein metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:345-57. [PMID: 21736953 DOI: 10.1016/j.bbalip.2011.06.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/01/2011] [Accepted: 06/14/2011] [Indexed: 12/13/2022]
Abstract
The understanding of the physiological and pathophysiological role of PLTP has greatly increased since the discovery of PLTP more than a quarter of century ago. A comprehensive review of PLTP is presented on the following topics: PLTP gene organization and structure; PLTP transfer properties; different forms of PLTP; characteristics of plasma PLTP complexes; relationship of plasma PLTP activity, mass and specific activity with lipoprotein and metabolic factors; role of PLTP in lipoprotein metabolism; PLTP and reverse cholesterol transport; insights from studies of PLTP variants; insights of PLTP from animal studies; PLTP and atherosclerosis; PLTP and signal transduction; PLTP in the brain; and PLTP in human disease. PLTP's central role in lipoprotein metabolism and lipid transport in the vascular compartment has been firmly established. However, more studies are needed to further delineate PLTP's functions in specific tissues, such as the lung, brain and adipose tissue. Furthermore, the specific role that PLTP plays in human diseases, such as atherosclerosis, cancer, or neurodegenerative disease, remains to be clarified. Exciting directions for future research include evaluation of PLTP's physiological relevance in intracellular lipid metabolism and signal transduction, which undoubtedly will advance our knowledge of PLTP functions in health and disease. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).
Collapse
Affiliation(s)
- John J Albers
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, 401 Queen Anne Ave N, Seattle, WA 98109, USA.
| | | | | |
Collapse
|
38
|
Johansen CT, Kathiresan S, Hegele RA. Genetic determinants of plasma triglycerides. J Lipid Res 2010; 52:189-206. [PMID: 21041806 DOI: 10.1194/jlr.r009720] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Plasma triglyceride (TG) concentration is reemerging as an important cardiovascular disease risk factor. More complete understanding of the genes and variants that modulate plasma TG should enable development of markers for risk prediction, diagnosis, prognosis, and response to therapies and might help specify new directions for therapeutic interventions. Recent genome-wide association studies (GWAS) have identified both known and novel loci associated with plasma TG concentration. However, genetic variation at these loci explains only ∼10% of overall TG variation within the population. As the GWAS approach may be reaching its limit for discovering genetic determinants of TG, alternative genetic strategies, such as rare variant sequencing studies and evaluation of animal models, may provide complementary information to flesh out knowledge of clinically and biologically important pathways in TG metabolism. Herein, we review genes recently implicated in TG metabolism and describe how some of these genes likely modulate plasma TG concentration. We also discuss lessons regarding plasma TG metabolism learned from various genomic and genetic experimental approaches. Treatment of patients with moderate to severe hypertriglyceridemia with existing therapies is often challenging; thus, gene products and pathways found in recent genetic research studies provide hope for development of more effective clinical strategies.
Collapse
Affiliation(s)
- Christopher T Johansen
- Department of Biochemistry, Robarts Research Institute, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | | | | |
Collapse
|
39
|
Liu HR, Wu G, Zhou B, Chen BS. Low cholesteryl ester transfer protein and phospholipid transfer protein activities are the factors making tree shrew and beijing duck resistant to atherosclerosis. Lipids Health Dis 2010; 9:114. [PMID: 20937151 PMCID: PMC2964723 DOI: 10.1186/1476-511x-9-114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 10/12/2010] [Indexed: 11/20/2022] Open
Abstract
Background Tree shrew and beijing duck are regarded as animal models resistant to atherosclerosis (AS). This study was carried out to discover the potential mechanism. Methods Blood samples were collected from healthy men and male animals. Plasma lipid profile and activities of cholesteryl ester transfer protein (CETP) and phospholipid transfer protein (PLTP) were measured, compared and analyzed in human, tree shrew, and Beijing duck. Results The results showed that there were species differences on plasma lipid profile and activities of CETP and PLTP in the three species. Compared with human, tree shrew and beijing duck had higher high density lipoprotein cholesterol (HDL-C)/total cholesterol (TC) and HDL-C/low density lipoprotein cholesterol (LDL-C) ratios, but lower CETP and PLTP activities. In the three species, CETP and PLTP activities were negatively related with the ratio of HDL-C/LDL-C. Conclusions The present study suggested that low plasma CETP and PLTP activities may lead to a high HDL-C/LDL-C ratio and a high resistance to AS finally in tree shrew and beijing duck. Moreover, low PLTP activity may also make the animals resistant to AS by the relative high vitamin E content of apoB-containing lipoproteins and high anti-inflammatory and antioxidative properties of HDL particles. A detailed study in the future is recommended.
Collapse
Affiliation(s)
- Hui-rong Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China.
| | | | | | | |
Collapse
|
40
|
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
|
41
|
Luo Y, Shelly L, Sand T, Chang G, Jiang XC. Identification and characterization of dual inhibitors for phospholipid transfer protein and microsomal triglyceride transfer protein. J Pharmacol Exp Ther 2010; 335:653-8. [PMID: 20805304 DOI: 10.1124/jpet.110.171942] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phospholipid transfer protein (PLTP) plays an important role in atherogenesis and lipoprotein metabolism. PLTP exerts its functions intracellularly and extracellularly. Both PLTP and microsomal triglyceride transfer protein (MTP) have been shown to regulate the secretion of apolipoprotein B (apoB) in hepatocytes. We have previously reported the characterization of inhibitors that selectively inhibit PLTP activity and reduce apoB secretion in hepatocytes. In the present study, we identified more compounds that inhibit both PLTP and MTP activity to various extents. These dual inhibitors are structurally different from the PLTP-selective inhibitors. In human hepatoma cell lines, dual inhibitors seem to be more effective in reducing apoB secretion than selective PLTP or MTP inhibitors. Furthermore, the dual inhibitors markedly reduced triglyceride secretion from hepatocytes. In the absence of PLTP, the dual inhibitors can further reduce apoB secretion, whereas selective PLTP inhibitors had no effect. We conclude that MTP and PLTP may work coordinately in the process of hepatic apoB assembly and secretion. To avoid liver toxicity mediated by MTP inhibition, selective PLTP inhibitors should be pursued.
Collapse
Affiliation(s)
- Yi Luo
- Department of Cardiovascular Metabolic and Endocrine Disease, Pfizer Global Research Division, Pfizer Inc, Groton,Connecticut 06340, USA.
| | | | | | | | | |
Collapse
|
42
|
Trevaskis NL, Shanker RM, Charman WN, Porter CJH. The Mechanism of Lymphatic Access of Two Cholesteryl Ester Transfer Protein Inhibitors (CP524,515 and CP532,623) and Evaluation of Their Impact on Lymph Lipoprotein Profiles. Pharm Res 2010; 27:1949-64. [DOI: 10.1007/s11095-010-0199-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 06/16/2010] [Indexed: 10/19/2022]
|
43
|
Bäckhed F. 99th Dahlem conference on infection, inflammation and chronic inflammatory disorders: the normal gut microbiota in health and disease. Clin Exp Immunol 2010; 160:80-4. [PMID: 20415855 DOI: 10.1111/j.1365-2249.2010.04123.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mammals are metagenomic, in that they are composed not only of their own genome but also those of all of their associated microbes (microbiome). Individual variations in the microbiome influence host health and may be implicated in disease aetiology. Therefore, it is not surprising that decreased microbial diversity is associated with both obesity and inflammatory bowel disease. Studies in germ-free mice have demonstrated that the gut microbiota is required for development of diet-induced obesity as well as inflammatory diseases. However, the underlying molecular mechanism(s) for how the gut microbiota causes metabolic diseases is only beginning to be clarified. Furthermore, emerging data suggest that the gut microbiota may predispose or protect against other important diseases such as cardiovascular disease and diabetes.
Collapse
Affiliation(s)
- F Bäckhed
- Sahlgrenska Center for Cardiovascular and Metabolic Research/Wallenberg Laboratory, Göteborg, Sweden.
| |
Collapse
|
44
|
Trevaskis NL, McEvoy CL, McIntosh MP, Edwards GA, Shanker RM, Charman WN, Porter CJH. The Role of the Intestinal Lymphatics in the Absorption of Two Highly Lipophilic Cholesterol Ester Transfer Protein Inhibitors (CP524,515 and CP532,623). Pharm Res 2010; 27:878-93. [DOI: 10.1007/s11095-010-0083-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 02/09/2010] [Indexed: 01/01/2023]
|
45
|
Luo Y, Shelly L, Sand T, Reidich B, Chang G, Macdougall M, Peakman MC, Jiang XC. Pharmacologic inhibition of phospholipid transfer protein activity reduces apolipoprotein-B secretion from hepatocytes. J Pharmacol Exp Ther 2009; 332:1100-6. [PMID: 19933370 DOI: 10.1124/jpet.109.161232] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phospholipid transfer protein (PLTP) plays an important role in atherogenesis, and its function goes well beyond that of transferring phospholipids between lipoprotein particles. Previous studies showed that genetic deficiency of PLTP in mice causes a substantially impaired hepatic secretion of apolipoprotein-B (apoB), the major protein of atherogenic lipoproteins. To understand whether the impaired apoB secretion is a direct result from lack of PLTP activity, in this study, we further investigated the function of PLTP in apoB secretion by using PLTP inhibitors. We identified a series of compounds containing a 3-benzazepine core structure that inhibit PLTP activity. Compound A, the most potent inhibitor, was characterized further and had little cross-reactivity with microsomal triglyceride transfer protein. Compound A reduced apoB secretion in human hepatoma cell lines and mouse primary hepatocytes. Furthermore, we confirmed that the reduction of apoB secretion mediated by compound A is PLTP-dependent, because the PLTP inhibitor had no effect on apoB secretion from PLTP-deficient hepatocytes. These studies provided evidence that PLTP activity regulates apoB secretion and pharmacologic inhibition of PLTP may be a new therapy for dyslipidemia by reducing apoB secretion.
Collapse
Affiliation(s)
- Yi Luo
- Department of Cardiovascular and Metabolic Diseases, Pfizer Global Research Division, Pfizer Inc., Groton, CT 06340, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Li Z, Li Y, Chakraborty M, Fan Y, Bui HH, Peake DA, Kuo MS, Xiao X, Cao G, Jiang XC. Liver-specific deficiency of serine palmitoyltransferase subunit 2 decreases plasma sphingomyelin and increases apolipoprotein E levels. J Biol Chem 2009; 284:27010-9. [PMID: 19648608 DOI: 10.1074/jbc.m109.042028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Sphingomyelin (SM) is one of the major lipid components of plasma lipoproteins. Serine palmitoyltransferase (SPT) is the key enzyme in SM biosynthesis. Mice totally lacking in SPT are embryonic lethal. The liver is the major site for plasma lipoprotein biosynthesis, secretion, and degradation, and in this study we utilized a liver-specific knock-out approach for evaluating liver SPT activity and also its role in plasma SM and lipoprotein metabolism. We found that a deficiency of liver-specific Sptlc2 (a subunit of SPT) decreased liver SPT protein mass and activity by 95 and 92%, respectively, but had no effect on other tissues. Liver Sptlc2 deficiency decreased plasma SM levels (in both high density lipoprotein and non-high density lipoprotein fractions) by 36 and 35% (p < 0.01), respectively, and increased phosphatidylcholine levels by 19% (p < 0.05), thus increasing the phosphatidylcholine/SM ratio by 77% (p < 0.001), compared with controls. This deficiency also decreased SM levels in the liver by 38% (p < 0.01) and in the hepatocyte plasma membranes (based on a lysenin-mediated cell lysis assay). Liver-specific Sptlc2 deficiency significantly increased hepatocyte apoE secretion and thus increased plasma apoE levels 3.5-fold (p < 0.0001). Furthermore, plasma from Sptlc2 knock-out mice had a significantly stronger potential for promoting cholesterol efflux from macrophages than from wild-type mice (p < 0.01) because of a greater amount of apoE in the circulation. As a result of these findings, we believe that the ability to control liver SPT activity could result in regulation of lipoprotein metabolism and might have an impact on the development of atherosclerosis.
Collapse
Affiliation(s)
- Zhiqiang Li
- State University of New York Downstate Medical Center, Brooklyn, New York 11203, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tzotzas T, Desrumaux C, Lagrost L. Plasma phospholipid transfer protein (PLTP): review of an emerging cardiometabolic risk factor. Obes Rev 2009; 10:403-11. [PMID: 19413703 DOI: 10.1111/j.1467-789x.2009.00586.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Plasma phospholipid transfer protein (PLTP) is a lipid transfer glycoprotein that binds to and transfers a number of amphipathic compounds. In earlier studies, the attention of the scientific community focused on the positive role of PLTP in high-density lipoprotein (HDL) metabolism. However, this potentially anti-atherogenic role of PLTP has been challenged recently by another picture: PLTP arose as a pro-atherogenic factor through its ability to increase the production of apolipoprotein B-containing lipoproteins, to decrease their antioxidative protection and to trigger inflammation. In humans, PLTP has mostly been studied in patients with cardiometabolic disorders. Both PLTP and related cholesteryl ester transfer protein (CETP) are secreted proteins, and adipose tissue is an important contributor to the systemic pools of these two proteins. Coincidently, high levels of PLTP and CETP have been found in the plasma of obese patients. PLTP activity and mass have been reported to be abnormally elevated in type 2 diabetes mellitus (T2DM) and insulin-resistant states, and this elevation is frequently associated with hypertriglyceridemia and obesity. This review article presents the state of knowledge on the implication of PLTP in lipoprotein metabolism, on its atherogenic potential, and the complexity of its implication in obesity, insulin resistance and T2DM.
Collapse
Affiliation(s)
- T Tzotzas
- Department of Nutrition and Dietetics, Technological Educational Institution, Thessaloniki, Greece.
| | | | | |
Collapse
|
48
|
Reverse modulation of the HDL Anionic Peptide Factor and phospholipid transfer protein activity in coronary artery disease and type 2 diabetes mellitus. Clin Biochem 2009; 42:845-51. [DOI: 10.1016/j.clinbiochem.2008.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 12/10/2008] [Accepted: 12/11/2008] [Indexed: 11/22/2022]
|
49
|
Ataxia with vitamin E deficiency with a mutation in a phospholipid transfer protein gene. J Neurol 2009; 256:1180-1. [PMID: 19252769 DOI: 10.1007/s00415-009-5075-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2008] [Revised: 01/23/2009] [Accepted: 02/12/2009] [Indexed: 02/06/2023]
|
50
|
Engler MB, Pullinger CR, Malloy MJ, Natanzon Y, Kulkarni MV, Song J, Eng C, Huuskonen J, Rivera C, Poon A, Bensley M, Sehnert A, Zellner C, Kane J, Aouizerat BE. Genetic variation in phospholipid transfer protein modulates lipoprotein profiles in hyperalphalipoproteinemia. Metabolism 2008; 57:1719-24. [PMID: 19013296 PMCID: PMC2615231 DOI: 10.1016/j.metabol.2008.07.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Accepted: 07/17/2008] [Indexed: 02/07/2023]
Abstract
We previously demonstrated the role of a phospholipid transfer protein (PLTP) gene variation (rs2294213) in determining levels of high-density lipoprotein cholesterol (HDL-C) in hypoalphalipoproteinemia (HypoA). We have now explored the role of PLTP in hyperalphalipoproteinemia (HyperA). The human PLTP gene was screened for sequence anomalies by DNA melting in 107 subjects with HyperA. The association with plasma lipoprotein levels was evaluated. We detected 7 sequence variations: 1 previously reported variation (rs2294213) and 5 novel mutations including 1 missense mutation (L106F). The PLTP activity was unchanged in the p.L106F mutation. The frequency of the rs2294213 minor allele was markedly increased in the HyperA group (7.0%) in comparison with a control group (4.3%) and the hypoalphalipoproteinemia group (2.2%). Moreover, rs2294213 was strongly associated with HDL-C levels. Linear regression models predict that possession of the rs2294213 minor allele increases HDL-C independent of triglycerides. These findings extend the association of rs2294213 with HDL-C levels into the extremes of the HDL distribution.
Collapse
Affiliation(s)
- Mary B. Engler
- Department of Physiological Nursing, University of California San Francisco, San Francisco, CA 94143
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143
| | - Clive R. Pullinger
- Department of Physiological Nursing, University of California San Francisco, San Francisco, CA 94143
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Mary J. Malloy
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Yanina Natanzon
- Department of Physiological Nursing, University of California San Francisco, San Francisco, CA 94143
| | - Medha V. Kulkarni
- Department of Physiological Nursing, University of California San Francisco, San Francisco, CA 94143
| | - James Song
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Celeste Eng
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Jaarko Huuskonen
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Christopher Rivera
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Annie Poon
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Matt Bensley
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143
| | - Amy Sehnert
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143
| | - Christian Zellner
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - John Kane
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Bradley E. Aouizerat
- Department of Physiological Nursing, University of California San Francisco, San Francisco, CA 94143
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143
| |
Collapse
|