1
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
2
|
Na AY, Lee H, Min EK, Paudel S, Choi SY, Sim H, Liu KH, Kim KT, Bae JS, Lee S. Novel Time-dependent Multi-omics Integration in Sepsis-associated Liver Dysfunction. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1101-1116. [PMID: 37084954 PMCID: PMC11082264 DOI: 10.1016/j.gpb.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/03/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
The recently developed technologies that allow the analysis of each single omics have provided an unbiased insight into ongoing disease processes. However, it remains challenging to specify the study design for the subsequent integration strategies that can associate sepsis pathophysiology and clinical outcomes. Here, we conducted a time-dependent multi-omics integration (TDMI) in a sepsis-associated liver dysfunction (SALD) model. We successfully deduced the relation of the Toll-like receptor 4 (TLR4) pathway with SALD. Although TLR4 is a critical factor in sepsis progression, it is not specified in single-omics analyses but only in the TDMI analysis. This finding indicates that the TDMI-based approach is more advantageous than single-omics analyses in terms of exploring the underlying pathophysiological mechanism of SALD. Furthermore, TDMI-based approach can be an ideal paradigm for insightful biological interpretations of multi-omics datasets that will potentially reveal novel insights into basic biology, health, and diseases, thus allowing the identification of promising candidates for therapeutic strategies.
Collapse
Affiliation(s)
- Ann-Yae Na
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyojin Lee
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Eun Ki Min
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Sanjita Paudel
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - So Young Choi
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - HyunChae Sim
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kwang-Hyeon Liu
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ki-Tae Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Jong-Sup Bae
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sangkyu Lee
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
3
|
Liao YE, Liu J, Arnold K. Heparan sulfates and heparan sulfate binding proteins in sepsis. Front Mol Biosci 2023; 10:1146685. [PMID: 36865384 PMCID: PMC9971734 DOI: 10.3389/fmolb.2023.1146685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Heparan sulfates (HSs) are the main components in the glycocalyx which covers endothelial cells and modulates vascular homeostasis through interactions with multiple Heparan sulfate binding proteins (HSBPs). During sepsis, heparanase increases and induces HS shedding. The process causes glycocalyx degradation, exacerbating inflammation and coagulation in sepsis. The circulating heparan sulfate fragments may serve as a host defense system by neutralizing dysregulated Heparan sulfate binding proteins or pro-inflammatory molecules in certain circumstances. Understanding heparan sulfates and heparan sulfate binding proteins in health and sepsis is critical to decipher the dysregulated host response in sepsis and advance drug development. In this review, we will overview the current understanding of HS in glycocalyx under septic condition and the dysfunctional heparan sulfate binding proteins as potential drug targets, particularly, high mobility group box 1 (HMGB1) and histones. Moreover, several drug candidates based on heparan sulfates or related to heparan sulfates, such as heparanase inhibitors or heparin-binding protein (HBP), will be discussed regarding their recent advances. By applying chemical or chemoenzymatic approaches, the structure-function relationship between heparan sulfates and heparan sulfate binding proteins is recently revealed with structurally defined heparan sulfates. Such homogenous heparan sulfates may further facilitate the investigation of the role of heparan sulfates in sepsis and the development of carbohydrate-based therapy.
Collapse
Affiliation(s)
- Yi-En Liao
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | | |
Collapse
|
4
|
Ruiz-Sanmartín A, Ribas V, Suñol D, Chiscano-Camón L, Palmada C, Bajaña I, Larrosa N, González JJ, Canela N, Ferrer R, Ruiz-Rodríguez JC. Characterization of a proteomic profile associated with organ dysfunction and mortality of sepsis and septic shock. PLoS One 2022; 17:e0278708. [PMID: 36459524 PMCID: PMC9718383 DOI: 10.1371/journal.pone.0278708] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
INTRODUCTION The search for new biomarkers that allow an early diagnosis in sepsis and predict its evolution has become a necessity in medicine. The objective of this study is to identify, through omics techniques, potential protein biomarkers that are expressed in patients with sepsis and their relationship with organ dysfunction and mortality. METHODS Prospective, observational and single-center study that included adult patients (≥ 18 years) who were admitted to a tertiary hospital and who met the criteria for sepsis. A mass spectrometry-based approach was used to analyze the plasma proteins in the enrolled subjects. Subsequently, using recursive feature elimination classification and cross-validation with a vector classifier, an association of these proteins with mortality and organ dysfunction was established. The protein-protein interaction network was analyzed with String software. RESULTS 141 patients were enrolled in this study. Mass spectrometry identified 177 proteins. Of all of them, and by recursive feature elimination, nine proteins (GPX3, APOB, ORM1, SERPINF1, LYZ, C8A, CD14, APOC3 and C1QC) were associated with organ dysfunction (SOFA > 6) with an accuracy of 0.82 ± 0.06, precision of 0.85 ± 0.093, sensitivity 0.81 ± 0.10, specificity 0.84 ± 0.10 and AUC 0.82 ± 0.06. Twenty-two proteins (CLU, LUM, APOL1, SAA1, CLEBC3B, C8A, ITIH4, KNG1, AGT, C7, SAA2, APOH, HRG, AFM, APOE, APOC1, C1S, SERPINC1, IGFALS, KLKB1, CFB and BTD) were associated with mortality with an accuracy of 0.86 ± 0.05, a precision of 0.91 ± 0.05, a sensitivity of 0.91 ± 0.05, a specificity of 0.72 ± 0.17, and an area under the curve (AUC) of 0.81 ± 0.08 with a confidence interval of 95%. CONCLUSION In sepsis there are proteomic patterns associated with organ dysfunction and mortality.
Collapse
Affiliation(s)
- Adolfo Ruiz-Sanmartín
- Department of Intensive Care, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vicent Ribas
- Eurecat, Centre Tecnològic de Catalunya, Digital Health Unit, Barcelona, Spain
| | - David Suñol
- Eurecat, Centre Tecnològic de Catalunya, Digital Health Unit, Barcelona, Spain
| | - Luis Chiscano-Camón
- Department of Intensive Care, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Clara Palmada
- Department of Intensive Care, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Iván Bajaña
- Department of Intensive Care, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Nieves Larrosa
- Department of Clinical Microbiology, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBERINFEC, ISCIII–CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan José González
- Department of Clinical Microbiology, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBERINFEC, ISCIII–CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Núria Canela
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Reus, Spain
| | - Ricard Ferrer
- Department of Intensive Care, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Carlos Ruiz-Rodríguez
- Department of Intensive Care, Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
5
|
Saito A, Ishimori N, Tokuhara S, Homma T, Nishikawa M, Iwabuchi K, Tsutsui H. Activation of Invariant Natural Killer T Cells by α-Galactosylceramide Attenuates the Development of Angiotensin II-Mediated Abdominal Aortic Aneurysm in Obese ob/ob Mice. Front Cardiovasc Med 2021; 8:659418. [PMID: 34041282 PMCID: PMC8141584 DOI: 10.3389/fcvm.2021.659418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/13/2021] [Indexed: 11/13/2022] Open
Abstract
The infiltration and activation of macrophages as well as lymphocytes within the aorta contribute to the pathogenesis of abdominal aortic aneurysm (AAA). Invariant natural killer T (iNKT) cells are unique subset of T lymphocytes and have a crucial role in atherogenesis. However, it remains unclear whether iNKT cells also impact on the development of AAA. Ob/ob mice were administered angiotensin II (AngII, 1,000 ng/kg/min) or phosphate-buffered saline (PBS) by osmotic minipumps for 4 weeks and further divided into 2 groups; α-galactosylceramide (αGC; PBS-αGC; n = 5 and AngII-αGC; n = 12), which specifically activates iNKT cells, and PBS (PBS-PBS; n = 10, and AngII-PBS; n = 6). Maximal abdominal aortic diameter was comparable between PBS-PBS and PBS-αGC, and was significantly greater in AngII-PBS than in PBS-PBS. This increase was significantly attenuated in AngII-αGC without affecting blood pressure. αGC significantly enhanced iNKT cell infiltration compared to PBS-PBS. The ratio of F4/80-positive macrophages or CD3-positive T lymphocytes area to the lesion area was significantly higher in AngII-PBS than in PBS-PBS, and was significantly decreased in AngII-αGC. Gene expression of M2-macrophage specific markers, arginase-1 and resistin-like molecule alpha, was significantly greater in aortic tissues from AngII-αGC compared to AngII-PBS 1 week after AngII administration, and this increase was diminished at 4 weeks. Activation of iNKT cells by αGC can attenuate AngII-mediated AAA in ob/ob mice via inducing anti-inflammatory M2 polarized state. Activation of iNKT cells by the bioactive lipid αGC may be a novel therapeutic target against the development of AAA.
Collapse
Affiliation(s)
- Akimichi Saito
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoki Ishimori
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Tokuhara
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tsuneaki Homma
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mikito Nishikawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuya Iwabuchi
- Department of Immunology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| |
Collapse
|
6
|
Wang H, Du S, Cai J, Wang J, Shen X. Apolipoprotein E2 Promotes the Migration and Invasion of Pancreatic Cancer Cells via Activation of the ERK1/2 Signaling Pathway. Cancer Manag Res 2020; 12:13161-13171. [PMID: 33376407 PMCID: PMC7764636 DOI: 10.2147/cmar.s284115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Background Apolipoprotein E2 (ApoE2) is reported to be essential for cell metastasis and proliferation and has been considered a potential diagnostic marker in many cancers. However, the function of ApoE2 in the metastasis of pancreatic cancer, as well as the underlying mechanism, remain unclear. Purpose In this study, we explored the effect of ApoE2 on the migration and invasion abilities of pancreatic cancer cells and explored the underlying molecular mechanism. Methods and Results Wound healing and Matrigel Transwell assays were used to investigate the role of ApoE2 in cell migration and invasion. Western blotting analysis showed that ApoE2 was overexpressed in pancreatic cancer tissues. Additionally, the overexpression of ApoE2 promoted the process of epithelial–mesenchymal transition (EMT) and enhanced the expression of MMP-2/9 in pancreatic cancer cells. Mechanistically, we found that inhibition of ERK1/2 signaling with PD98059 impaired the ApoE2-mediated promotion of cell migration, invasion and EMT. Conclusion This study demonstrated that ApoE2/ERK1/2 signaling promoted the migration and invasion of pancreatic cancer cells. ApoE2 might be a potential therapeutic target for the treatment of pancreatic cancer metastasis.
Collapse
Affiliation(s)
- Hui Wang
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Shaoxia Du
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Jun Cai
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Juan Wang
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Xiaohong Shen
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
7
|
Shao Y, Zhao T, Zhang W, He J, Lu F, Cai Y, Lai Z, Wei N, Liang C, Liu L, Hong Y, Cheng X, Li J, Tang P, Fan W, Ou M, Yang J, Liu Y, Cui L. Presence of the apolipoprotein E-ε4 allele is associated with an increased risk of sepsis progression. Sci Rep 2020; 10:15735. [PMID: 32978453 PMCID: PMC7519096 DOI: 10.1038/s41598-020-72616-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/08/2020] [Indexed: 12/15/2022] Open
Abstract
Growing evidence indicated that single nucleotide polymorphisms (SNPs) in the apolipoprotein E (APOE) gene are related to increase the risk of many inflammatory-related diseases. However, few genetic studies have associated the APOE gene polymorphism with sepsis. This study was to investigate the clinical relevance of the APOE gene polymorphism in the onset and progression of sepsis. A multicenter case–control association study with a large sample size (601 septic patients and 699 healthy individuals) was conducted. Clinical data showed that the APOEε4 allele was overrepresented among all patients with septic shock (p = 0.031) compared with sepsis subtype, suggesting that APOEε4 allele may associated with increased susceptibility to the progression of sepsis. Moreover, the APOE mRNA levels decreased after lipopolysaccharide (LPS) stimulation in cells in culture. Then 21 healthy individuals to extract PBMC for genotype grouping (APOE4+ group 8; APOE4− group 13) was selected to evaluate the effect on APOE level, and results showed that the expression level of APOE in APOE4+ group and APOE4− group did not differ in mRNA levels after an LPS challenge, but the protein levels in APOE4+ group decreased slower than that in APOE4− group, and this process was accompanied by the upregulation of proinflammatory cytokines. These results provide evidence that the APOEε4 allele might be associated with the development of sepsis and a potential risk factor that can be used in the prognosis of sepsis.
Collapse
Affiliation(s)
- Yiming Shao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China.,The Intensive Care Unit, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China.,Zhanjiang Key Laboratory of Organ Injury and Protection and Translational Medicine, Guangdong, 524000, China
| | - Tian Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China.,School of Clinical Medicine, Hubei University of Science and Technology, Xianning, 437000, China
| | - Wenying Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Junbing He
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-Sen University, Jieyang, 52200, China
| | - Furong Lu
- The Intensive Care Unit, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China.,The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-Sen University, Jieyang, 52200, China
| | - Yujie Cai
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Zhipeng Lai
- The Intensive Care Unit, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Ning Wei
- The Intensive Care Unit, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Chunmei Liang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Lizhen Liu
- The Intensive Care Unit, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Yuan Hong
- The Intensive Care Unit, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Xiaohong Cheng
- The Intensive Care Unit, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Jia Li
- The Intensive Care Unit ,The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Pei Tang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Weihao Fan
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Mingqian Ou
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Jingqi Yang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Yansong Liu
- The Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China.
| |
Collapse
|
8
|
Bathini P, Brai E, Auber LA. Olfactory dysfunction in the pathophysiological continuum of dementia. Ageing Res Rev 2019; 55:100956. [PMID: 31479764 DOI: 10.1016/j.arr.2019.100956] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/29/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022]
Abstract
Sensory capacities like smell, taste, hearing, vision decline with aging, but increasing evidence show that sensory dysfunctions are one of the early signs diagnosing the conversion from physiological to pathological brain state. Smell loss represents the best characterized sense in clinical practice and is considered as one of the first preclinical signs of Alzheimer's and Parkinson's disease, occurring a decade or more before the onset of cognitive and motor symptoms. Despite the numerous scientific reports and the adoption in clinical practice, the etiology of sensory damage as prodromal of dementia remains largely unexplored and more studies are needed to resolve the mechanisms underlying sensory network dysfunction. Although both cognitive and sensory domains are progressively affected, loss of sensory experience in early stages plays a major role in reducing the autonomy of demented people in their daily tasks or even possibly contributing to their cognitive decline. Interestingly, the chemosensory circuitry is devoid of a blood brain barrier, representing a vulnerable port of entry for neurotoxic species that can spread to the brain. Furthermore, the exposure of the olfactory system to the external environment make it more susceptible to mechanical injury and trauma, which can cause degenerative neuroinflammation. In this review, we will summarize several findings about chemosensory impairment signing the conversion from healthy to pathological brain aging and we will try to connect those observations to the promising research linking environmental influences to sporadic dementia. The scientific body of knowledge will support the use of chemosensory diagnostics in the presymptomatic stages of AD and other biomarkers with the scope of finding treatment strategies before the onset of the disease.
Collapse
Affiliation(s)
- Praveen Bathini
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Emanuele Brai
- VIB-KU Leuven Center for Brain & Disease Research, Laboratory for the Research of Neurodegenerative Diseases, Leuven, Belgium
| | - Lavinia Alberi Auber
- Department of Medicine, University of Fribourg, Fribourg, Switzerland; Swiss Integrative Center of Human Health, Fribourg, Switzerland.
| |
Collapse
|
9
|
Gordon EM, Yao X, Xu H, Karkowsky W, Kaler M, Kalchiem-Dekel O, Barochia AV, Gao M, Keeran KJ, Jeffries KR, Levine SJ. Apolipoprotein E is a concentration-dependent pulmonary danger signal that activates the NLRP3 inflammasome and IL-1β secretion by bronchoalveolar fluid macrophages from asthmatic subjects. J Allergy Clin Immunol 2019; 144:426-441.e3. [PMID: 30872118 DOI: 10.1016/j.jaci.2019.02.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 02/15/2019] [Accepted: 02/26/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND House dust mite (HDM)-challenged Apoe-/- mice display enhanced airway hyperreactivity and mucous cell metaplasia. OBJECTIVE We sought to characterize the pathways that induce apolipoprotein E (APOE) expression by bronchoalveolar lavage fluid (BALF) macrophages from asthmatic subjects and identify how APOE regulates IL-1β secretion. METHODS Macrophages were isolated from asthmatic BALF and derived from THP-1 cells and human monocytes. RESULTS HDM-derived cysteine and serine proteases induced APOE secretion from BALF macrophages through protease-activated receptor 2. APOE at concentrations of less than 2.5 nmol/L, which are similar to levels found in epithelial lining fluid from healthy adults, did not induce IL-1β release from BALF macrophages. In contrast, APOE at concentrations of 25 nmol/L or greater induced nucleotide-binding oligomerization domain, leucine-rich repeat-containing protein (NLRP) 3 and pro-IL-1β expression by BALF macrophages, as well as the caspase-1-mediated generation of mature IL-1β secreted from cells. HDM acted synergistically with APOE to both prime and activate the NLRP3 inflammasome. In a murine model of neutrophilic airway inflammation induced by HDM and polyinosinic-polycytidylic acid, APOE reached a concentration of 32 nmol/L in epithelial lining fluid, with associated increases in BALF IL-1β levels. APOE-dependent NLRP3 inflammasome activation in macrophages was primarily mediated through a potassium efflux-dependent mechanism. CONCLUSION APOE can function as an endogenous, concentration-dependent pulmonary danger signal that primes and activates the NLPR3 inflammasome in BALF macrophages from asthmatic subjects to secrete IL-1β. This might represent a mechanism through which APOE amplifies pulmonary inflammatory responses when concentrations in the lung are increased to greater than normal levels, which can occur during viral exacerbations of HDM-induced asthma characterized by neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Haitao Xu
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - William Karkowsky
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Or Kalchiem-Dekel
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Meixia Gao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Karen J Keeran
- Animal Surgery and Resources Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Kenneth R Jeffries
- Animal Surgery and Resources Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
10
|
LPS-Induced Systemic Inflammation Does Not Alter Atherosclerotic Plaque Area or Inflammation in APOE3∗LEIDEN Mice in the Early Phase Up to 15 Days. Shock 2018; 50:360-365. [DOI: 10.1097/shk.0000000000001026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Apolipoprotein E and Atherosclerosis: From Lipoprotein Metabolism to MicroRNA Control of Inflammation. J Cardiovasc Dev Dis 2018; 5:jcdd5020030. [PMID: 29789495 PMCID: PMC6023389 DOI: 10.3390/jcdd5020030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 12/26/2022] Open
Abstract
Apolipoprotein (apo) E stands out among plasma apolipoproteins through its unprecedented ability to protect against atherosclerosis. Although best recognized for its ability to mediate plasma lipoprotein clearance in the liver and protect against macrophage foam cell formation, our recent understanding of the influence that apoE can exert to control atherosclerosis has significantly widened. Among apoE’s newfound athero-protective properties include an ability to control exaggerated hematopoiesis, blood monocyte activation and aortic stiffening in mice with hyperlipidemia. Mechanisms responsible for these exciting new properties extend beyond apoE’s ability to prevent cellular lipid excess. Rather, new findings have revealed a role for apoE in regulating microRNA-controlled cellular signaling in cells of the immune system and vascular wall. Remarkably, infusions of apoE-responsive microRNA mimics were shown to substitute for apoE in protecting against systemic and vascular inflammation to suppress atherosclerosis in mice with hyperlipidemia. Finally, more recent evidence suggests that apoE may control the release of microvesicles that could modulate cellular signaling, inflammation and atherosclerosis at a distance. These exciting new findings position apoE within the emerging field of intercellular communication that could introduce new approaches to control atherosclerosis cardiovascular disease.
Collapse
|
12
|
Epigenetics, microbiota, and intraocular inflammation: New paradigms of immune regulation in the eye. Prog Retin Eye Res 2018; 64:84-95. [DOI: 10.1016/j.preteyeres.2018.01.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 01/07/2018] [Accepted: 01/11/2018] [Indexed: 01/15/2023]
|
13
|
Yao X, Gordon EM, Figueroa DM, Barochia AV, Levine SJ. Emerging Roles of Apolipoprotein E and Apolipoprotein A-I in the Pathogenesis and Treatment of Lung Disease. Am J Respir Cell Mol Biol 2017; 55:159-69. [PMID: 27073971 DOI: 10.1165/rcmb.2016-0060tr] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Emerging roles are being recognized increasingly for apolipoproteins in the pathogenesis and treatment of lung diseases on the basis of their ability to suppress inflammation, oxidative stress, and tissue remodeling, and to promote adaptive immunity and host defense. Apolipoproteins, such as apolipoprotein E (apoE) and apolipoprotein A-I (apoA-I), are important components of lipoprotein particles that facilitate the transport of cholesterol, triglycerides, and phospholipids between plasma and cells. ApoE-containing lipoprotein particles are internalized into cells by low-density lipoprotein receptors (LDLRs), whereas apoA-I can interact with the ATP-binding cassette subfamily A member 1 (ABCA1) transporter to efflux cholesterol and phospholipids out of cells. ApoE and apoA-I also mediate receptor-independent effects, such as binding to and neutralizing LPS. Both apoE and apoA-I are expressed by lung cells, which allows apoE/LDLR- and apoA-I/ABCA1-dependent pathways to modulate normal lung health and the pathogenesis of respiratory diseases, including asthma, acute lung injury, cancer, emphysema, pulmonary fibrosis, and pulmonary hypertension. Data from human studies and research using experimental murine model systems have shown that both apoE and apoA-I pathways play primarily protective roles in lung biology and respiratory disease. Furthermore, apolipoprotein mimetic peptides, corresponding to the LDLR-binding domain of apoE or the class A amphipathic α-helical structure of apoA-I, have antiinflammatory and antioxidant effects that attenuate the severity of lung disease in murine models. Thus, the development of inhaled apolipoprotein mimetic peptides as a novel treatment paradigm could represent a significant advance for patients with respiratory disease who do not respond to current therapies.
Collapse
Affiliation(s)
- Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Debbie M Figueroa
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
14
|
Walley KR, Francis GA, Opal SM, Stein EA, Russell JA, Boyd JH. The Central Role of Proprotein Convertase Subtilisin/Kexin Type 9 in Septic Pathogen Lipid Transport and Clearance. Am J Respir Crit Care Med 2016; 192:1275-86. [PMID: 26252194 DOI: 10.1164/rccm.201505-0876ci] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbial cell walls contain pathogenic lipids, including LPS in gram-negative bacteria, lipoteichoic acid in gram-positive bacteria, and phospholipomannan in fungi. These pathogen lipids are major ligands for innate immune receptors and figure prominently in triggering the septic inflammatory response. Alternatively, pathogen lipids can be cleared and inactivated, thus limiting the inflammatory response. Accordingly, biological mechanisms for sequestering and clearing pathogen lipids from the circulation have evolved. Pathogen lipids released into the circulation are initially bound by transfer proteins, notably LPS binding protein and phospholipid transfer protein, and incorporated into high-density lipoprotein particles. Next, LPS binding protein, phospholipid transfer protein, and other transfer proteins transfer these lipids to ApoB-containing lipoproteins, including low-density (LDL) and very-low-density lipoproteins and chylomicrons. Pathogen lipids within these lipoproteins and their remnants are then cleared from the circulation by the liver. Hepatic clearance involves the LDL receptor (LDLR) and possibly other receptors. Once absorbed by the liver, these lipids are then excreted in the bile. Recent evidence suggests pathogen lipid clearance can be modulated. Importantly, reduced proprotein convertase subtilisin/kexin type 9 activity increases recycling of the LDLR and thereby increases LDLR on the surface of hepatocytes, which increases clearance by the liver of pathogen lipids transported in LDL. Increased pathogen lipid clearance, which can be achieved by inhibiting proprotein convertase subtilisin/kexin type 9, may decrease the systemic inflammatory response to sepsis and improve clinical outcomes.
Collapse
Affiliation(s)
- Keith R Walley
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gordon A Francis
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven M Opal
- 2 Infectious Disease Division, Memorial Hospital of Rhode Island and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - Evan A Stein
- 3 Metabolic and Atherosclerosis Research Center, Cincinnati, Ohio
| | - James A Russell
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - John H Boyd
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
15
|
Szabo PA, Anantha RV, Shaler CR, McCormick JK, Haeryfar SMM. CD1d- and MR1-Restricted T Cells in Sepsis. Front Immunol 2015; 6:401. [PMID: 26322041 PMCID: PMC4533011 DOI: 10.3389/fimmu.2015.00401] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/22/2015] [Indexed: 12/23/2022] Open
Abstract
Dysregulated immune responses to infection, such as those encountered in sepsis, can be catastrophic. Sepsis is typically triggered by an overwhelming systemic response to an infectious agent(s) and is associated with high morbidity and mortality even under optimal critical care. Recent studies have implicated unconventional, innate-like T lymphocytes, including CD1d- and MR1-restricted T cells as effectors and/or regulators of inflammatory responses during sepsis. These cell types are typified by invariant natural killer T (iNKT) cells, variant NKT (vNKT) cells, and mucosa-associated invariant T (MAIT) cells. iNKT and vNKT cells are CD1d-restricted, lipid-reactive cells with remarkable immunoregulatory properties. MAIT cells participate in antimicrobial defense, and are restricted by major histocompatibility complex-related protein 1 (MR1), which displays microbe-derived vitamin B metabolites. Importantly, NKT and MAIT cells are rapid and potent producers of immunomodulatory cytokines. Therefore, they may be considered attractive targets during the early hyperinflammatory phase of sepsis when immediate interventions are urgently needed, and also in later phases when adjuvant immunotherapies could potentially reverse the dangerous state of immunosuppression. We will highlight recent findings that point to the significance or the therapeutic potentials of NKT and MAIT cells in sepsis and will also discuss what lies ahead in research in this area.
Collapse
Affiliation(s)
- Peter A Szabo
- Department of Microbiology and Immunology, Western University , London, ON , Canada
| | - Ram V Anantha
- Department of Microbiology and Immunology, Western University , London, ON , Canada ; Division of General Surgery, Department of Medicine, Western University , London, ON , Canada
| | - Christopher R Shaler
- Department of Microbiology and Immunology, Western University , London, ON , Canada
| | - John K McCormick
- Department of Microbiology and Immunology, Western University , London, ON , Canada ; Centre for Human Immunology, Western University , London, ON , Canada ; Lawson Health Research Institute , London, ON , Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University , London, ON , Canada ; Centre for Human Immunology, Western University , London, ON , Canada ; Lawson Health Research Institute , London, ON , Canada ; Division of Clinical Immunology and Allergy, Department of Medicine, Western University , London, ON , Canada
| |
Collapse
|
16
|
Borilova Linhartova P, Bartova J, Poskerova H, Machal J, Vokurka J, Fassmann A, Izakovicova Holla L. Apolipoprotein E gene polymorphisms in relation to chronic periodontitis, periodontopathic bacteria, and lipid levels. Arch Oral Biol 2015; 60:456-62. [DOI: 10.1016/j.archoralbio.2014.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/17/2014] [Accepted: 10/13/2014] [Indexed: 02/07/2023]
|
17
|
Zhang P, Li Y, Zhang LD, Wang LH, Wang X, He C, Lin ZF. Proteome changes in mesenteric lymph induced by sepsis. Mol Med Rep 2014; 10:2793-804. [PMID: 25242054 PMCID: PMC4227422 DOI: 10.3892/mmr.2014.2580] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 05/02/2014] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to examine the changes in mesenteric lymph during the development of sepsis and to identify the distinct proteins involved, as targets for further study. The sepsis animal model was constructed by cecal ligation and puncture (CLP). The mesenteric lymph was collected from 28 adult male Sprague-Dawley rats, which were randomly divided into the following four groups (n=7 per group): CLP-6 h, CLP-24 h, sham-6 h and sham-24 h groups. Capillary high performance liquid chromatography-tandem mass spectrometry was performed to analyze the proteome in mesenteric lymph. A comprehensive bioinformatic analysis was then conducted to investigate the distinct proteins. Compared with the sham group, 158 distinct proteins were identified in the lymph samples from the CLP group. Five of these proteins associated with the same lipid metabolism pathway were selected, apolipoprotein E (ApoE), annexin A1 (Anxa1), neutrophil gelatinase-associated lipocalin (NGAL), S100a8 and S100a9. The expression of ApoE, Anxa1, NGAL, S100a8 and S100a9 were all elevated in the progression of sepsis. The five proteins were reported to be closely associated with disease development and may be a potential target for the diagnosis and treatment of sepsis. In conclusion, identifying proteome changes in mesenteric lymph provides a novel perspective to understand the pathological mechanisms underlying sepsis.
Collapse
Affiliation(s)
- Ping Zhang
- Emergency Department, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yan Li
- Emergency Department, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai 201620, P.R. China
| | - Lian-Dong Zhang
- Emergency Department, Shuguang Hospital Baoshan Branch, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, P.R. China
| | - Liang-Hua Wang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, P.R. China
| | - Xi Wang
- Emergency Department, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Chao He
- Emergency Department, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Zhao-Fen Lin
- Emergency Department, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
18
|
Browne RW, Weinstock-Guttman B, Horakova D, Zivadinov R, Bodziak ML, Tamaño-Blanco M, Badgett D, Tyblova M, Vaneckova M, Seidl Z, Krasensky J, Bergsland N, Ramasamy DP, Hagemeier J, Havrdova E, Ramanathan M. Apolipoproteins are associated with new MRI lesions and deep grey matter atrophy in clinically isolated syndromes. J Neurol Neurosurg Psychiatry 2014; 85:859-64. [PMID: 24470599 DOI: 10.1136/jnnp-2013-307106] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES There is increasing evidence that serum lipoprotein cholesterol biomarkers are associated with disease progression in clinically isolated syndromes (CIS). Apolipoproteins (Apo) are recognition ligands that mediate the physiological interactions of cholesterol-containing lipoproteins. The objective of this study was to investigate whether serum Apo levels are associated with CIS disease progression. METHODS ApoB, ApoAI, ApoAII, ApoE and lipoprotein (a) (Lpa) levels were measured in serum samples obtained prior to the start of treatment from 181 CIS patients (123 women, 58 men, 68% women; mean age: 28.1±SD 8.1 years). All patients were treated with intramuscular interferon-β as part of the prospective study. Clinical and MRI assessments were obtained at baseline, 6, 12 and 24 months after start of interferon-β treatment. RESULTS Greater ApoB levels were associated with increased number of new T2 lesions (p<0.001) and increased number of new or enlarging T2 lesions (p<0.001) over 2 years. Each 10 mg/dL of greater baseline ApoB is associated with a 16% increase in the number of new T2 lesions over 2 years. ApoAI, ApoAII, ApoE and Lpa were not associated with T2 lesions. Greater ApoE levels were associated with greater deep grey matter atrophy (partial correlation rp=-0.28, p<0.001). Each 1 mg/dL increment in ApoE levels was associated with a 1% increase in deep grey matter atrophy over 2 years. CONCLUSIONS Serum ApoB levels are associated with new lesion accumulation whereas ApoE levels are associated with deep grey matter atrophy in high risk CIS patients treated with interferon β-1a.
Collapse
Affiliation(s)
- Richard W Browne
- Department of Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, New York, USA
| | | | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Robert Zivadinov
- Department of Neurology, State University of New York, Buffalo, New York, USA Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Mary Lou Bodziak
- Department of Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, New York, USA
| | - Miriam Tamaño-Blanco
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York, USA
| | - Darlene Badgett
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York, USA
| | - Michaela Tyblova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Zdenek Seidl
- Department of Radiology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Jan Krasensky
- Department of Radiology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Deepa P Ramasamy
- Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Jesper Hagemeier
- Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Eva Havrdova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Murali Ramanathan
- Department of Neurology, State University of New York, Buffalo, New York, USA Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York, USA
| |
Collapse
|
19
|
Cao Z, Yende S, Kellum JA, Angus DC, Robinson RAS. Proteomics reveals age-related differences in the host immune response to sepsis. J Proteome Res 2013; 13:422-32. [PMID: 24266763 DOI: 10.1021/pr400814s] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sepsis is commonly caused by community-acquired pneumonia (CAP) and may develop into severe sepsis, characterized by multiple organ failure. The risk of severe sepsis among CAP patients and subsequent mortality increases sharply after the age of 65. The molecular mechanisms associated with this age-related risk are not fully understood. To better understand factors involved with increased incidence and mortality of severe sepsis in the elderly, we used a nested case-control study of patients enrolled in a multicenter observational cohort of 2320 participants with CAP. We identified a total of 39 CAP patients 50-65 and 70-85 years old who did or did not develop severe sepsis. Plasma samples were obtained on presentation to the emergency department and prior to therapeutic interventions. A semiquantitative plasma proteomics workflow was applied which incorporated tandem immunoaffinity depletion, iTRAQ labeling, strong cation exchange fractionation, and nanoflow liquid chromatography coupled to high-resolution mass spectrometry. In total, 772 proteins were identified, of which 58 proteins exhibit statistically significant differences in expression levels among patients with severe sepsis as a function of age. Differentially expressed proteins are involved in pathways such as acute phase response, coagulation signaling, atherosclerosis signaling, lipid metabolism, and production of nitric oxide and reactive oxygen species. This study provides insight into factors that may explain age-related differences in incidence of severe sepsis in the elderly.
Collapse
Affiliation(s)
- Zhiyun Cao
- Department of Chemistry and ‡The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) Laboratory and Department of Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | | | | | | | | |
Collapse
|
20
|
Lv X, Ai J, Li M, Wang H, Chen T, Fang Y, Liu Y, Zhou P, Chen M, Tan R, Liu Y, Yang Y, Zhou Q. Comparative proteomics and correlated signaling network of kidney in ApoE
deficient mouse. Proteomics Clin Appl 2013; 7:829-38. [PMID: 23687078 DOI: 10.1002/prca.201200112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaoyan Lv
- Department of Dermatology, West China Hospital, West China Medical School; Sichuan University; Chengdu P. R. China
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Jianzhong Ai
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Mi Li
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Honglian Wang
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Tielin Chen
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Yin Fang
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Yunhong Liu
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Puhui Zhou
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Mianzhi Chen
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Ruizhi Tan
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Yuhang Liu
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Yang Yang
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| | - Qin Zhou
- Core Facility of Genetically Engineered Mice, West China Hospital, West China School of Medicine; Sichuan University; Chengdu P. R. China
| |
Collapse
|
21
|
Zhou TB. Signaling pathways of apoE and its role of gene expression in glomerulus diseases. J Recept Signal Transduct Res 2013; 33:73-78. [PMID: 23384034 DOI: 10.3109/10799893.2013.765466] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The roles of apolipoprotein E (apoE) in regulating plasma lipids and lipoproteins levels have been investigated for over several decades. However, in different tissues/cells, the role of apoE was different, such as that it was a risk factor for cancer, but some reports stated that apoE was a protective factor for renal diseases. At the moment, most of the studies find that apoE not only acts as a ligand for metabolism of lipids, but also plays as a factor to regulate lots of signaling pathways. There was rare review to sum up the signaling pathways for apoE, and there was also rare review to widely review the gene expression of apoE in glomerulus diseases. This review was performed to provide a relatively complete signaling pathways flowchart for apoE to the investigators who were interested in the roles of apoE in the pathogenesis of glomerulus diseases. In the past decades, some studies were also performed to explore the association of apoE gene expression with the risk of glomerulus diseases. However, the role of apoE in the pathogenesis of glomerulus diseases was controversial. Here, the signal transduction pathways of apoE and its role of gene expression in the pathogenesis of glomerulus diseases were reviewed.
Collapse
Affiliation(s)
- Tian-Biao Zhou
- Department of Pediatric Nephrology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China.
| |
Collapse
|
22
|
Monzon-Casanova E, Paletta D, Starick L, Müller I, Sant'Angelo DB, Pyz E, Herrmann T. Direct identification of rat iNKT cells reveals remarkable similarities to human iNKT cells and a profound deficiency in LEW rats. Eur J Immunol 2012; 43:404-15. [PMID: 23165932 DOI: 10.1002/eji.201242565] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 10/29/2012] [Accepted: 11/14/2012] [Indexed: 12/18/2022]
Abstract
iNKT cells are a particular lymphocyte population with potent immunomodulatory capa-city; by promoting or suppressing immune responses against infections, tumors, and autoimmunity, iNKT cells are a promising target for immunotherapy. The hallmark of iNKT cells is the expression of a semiinvariant TCR (with an invariant α-chain comprising AV14 and AJ18 gene segments), which recognizes glycolipids presented by CD1d. Here, we identified iNKT cells for the first time in the rat using rat CD1d-dimers and PLZF staining. Importantly, in terms of frequencies (1.05% ± 0.52 SD of all intrahepatic αβ T cells), coreceptor expression and in vitro expansion features, iNKT cells from F344 inbred rats more closely resemble human iNKT cells than their mouse counterparts. In contrast, in LEW inbred rats, which are often used as models for organ-specific autoimmune diseases, iNKT cell numbers are near or below the detection limit. Interestingly, the usage of members of the rat AV14 gene family differed between F344 and LEW inbred rats. In conclusion, the similarities between F344 rat and human iNKT cells and the nearly absent iNKT cells in LEW rats make the rat a promising animal model for the study of iNKT cell-based therapies and of iNKT-cell biology.
Collapse
Affiliation(s)
- Elisa Monzon-Casanova
- Institute for Virology and Immunobiology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Apolipoprotein (apo) E is a multifunctional protein that has long been recognized for its ability to safeguard against atherosclerosis. Among its pleiotropic roles known to suppress atherosclerosis, mechanisms by which apoE regulates cells of the immune system have remained elusive. Because atherosclerosis is a chronic inflammatory disease that remains on the rise, understanding in more detail how apoE controls immune cell activation and function is of much interest. RECENT FINDINGS Literature reported in the past year introduces apoE as a regulator of monocyte and macrophage plasticity. Through signals delivered by its interaction with cell surface receptors, apoE has been shown to influence the polarity and inflammatory phenotypes of the macrophage. By promoting cellular cholesterol efflux in a cell autonomous manner and through its ability to enhance HDL function in hyperlipidemic plasma, apoE is now known to suppress atherosclerosis by controlling myeloid cell proliferation, monocyte activation and their capacity to infiltrate the vascular wall. Lastly, the structural basis for apoE isoform-specific effects in macrophage dysfunction and atherosclerosis susceptibility is beginning to emerge. SUMMARY Collectively, these findings introduce a new dimension to our understanding of how apoE links lipoprotein biology to monocyte and macrophage function in atherosclerosis susceptibility.
Collapse
Affiliation(s)
- Robert L Raffai
- Department of Surgery, University of California San Francisco, and Veterans Affairs Medical Center, San Francisco, California 94121, USA.
| |
Collapse
|
24
|
Christoffersen C, Nielsen LB. Apolipoprotein M--a new biomarker in sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:126. [PMID: 22587809 PMCID: PMC3580609 DOI: 10.1186/cc11320] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sepsis is one of the leading causes of mortality in non-cardiac intensive care units, and the need for markers of progression and severity are high. Also, treatment of sepsis is highly debated and potential new targets of treatment are of great interest. In the previous issue of Critical Care Kumaraswamy and colleagues have investigated whether plasma apolipoprotein M (apoM) is affected during different grades of sepsis, septic shock and systemic inflammatory response syndrome. Interestingly, plasma apoM was significantly decreased in all groups of patients with a relationship to severity of disease. This identifies apoM as a potential new biomarker in sepsis. It also underscores the possibility that altered high-density lipoprotein in sepsis patients can affect the course of disease. Thus, since apoM is the carrier of Sphingosine-1-P (S1P), a molecule with great influence on vascular barrier function, the study presented raises the interest and relevance for further studies of apoM and S1P in relation to sepsis and inflammation.
Collapse
|
25
|
Datta G, Gupta H, Zhang Z, Mayakonda P, Anantharamaiah G, White CR. HDL Mimetic Peptide Administration Improves Left Ventricular Filling and Cardiac output in Lipopolysaccharide-Treated Rats. JOURNAL OF CLINICAL & EXPERIMENTAL CARDIOLOGY 2011; 2:1000172. [PMID: 23227448 PMCID: PMC3514969 DOI: 10.4172/2155-9880.1000172] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
AIMS: Cardiac dysfunction is a complication of sepsis and contributes to morbidity and mortality. Since raising plasma apolipoprotein (apo) A-I and high density lipoprotein (HDL) concentration reduces sepsis complications, we tested the hypothesis that the apoA-I mimetic peptide 4F confers similar protective effects in rats treated with lipopolysaccharide (LPS). METHODS AND RESULTS: Male Sprague-Dawley (SD) rats were randomized to receive saline vehicle (n=13), LPS (10 mg/kg: n=16) or LPS plus 4F (10 mg/kg each: n=13) by intraperitoneal injection. Plasma cytokine and chemokine levels were significantly elevated 24 hrs after LPS administration. Echocardiographic studies revealed changes in cardiac dimensions that resulted in a reduction in left ventricular end-diastolic volume (LVEDV), stroke volume (SV) and cardiac output (CO) 24 hrs after LPS administration. 4F treatment reduced plasma levels of inflammatory mediators and increased LV filling, resulting in improved cardiac performance. Chromatographic separation of lipoproteins from plasma of vehicle, LPS and LPS+4F rats revealed similar profiles. Further analyses showed that LPS treatment reduced the agarose electrophoretic mobility of isolated HDL fractions. HDL-associated proteins were characterized by SDSPAGE and mass spectrometry. ApoA-I and apoA-IV were reduced while apoE content was increased in LPStreated rats. 4F treatment in vivo attenuated changes in HDL-associated apolipoproteins and increased the electrophoretic mobility of the particle. CONCLUSIONS: The ability of 4F to reduce inflammation and improve cardiac performance in LPS-treated rats may be due to its capacity to neutralize endotoxin and prevent adverse changes in HDL composition and function.
Collapse
Affiliation(s)
- Geeta Datta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Himanshu Gupta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhenghao Zhang
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - G.M. Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C. Roger White
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
26
|
Wang C, Wang Y, Wang A, Fu P, Yang Y. The diagnostic value of apolipoprotein E in pediatric patients with invasive bacterial infections. Clin Biochem 2011; 45:215-8. [PMID: 22178110 DOI: 10.1016/j.clinbiochem.2011.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 11/11/2011] [Accepted: 11/14/2011] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate the potential diagnostic value of apolipoprotein E (ApoE) measurements in pediatric patients with invasive bacterial infections. DESIGN AND METHOD A total of 185 pediatric patients were enrolled in this study, including 94 patients with confirmable infections and 91 patients without confirmable infections. Serum and cerebrospinal fluid (CSF) ApoE levels were measured by immunoturbidimetry. The diagnostic values of ApoE were evaluated by the receiver operating curve (ROC) method. RESULTS ApoE levels in CSF were significantly increased in patients with bacterial meningitis, and serum ApoE was markedly elevated in patients with sepsis or with bacterial meningitis compared with patients with other infections and uninfected children. The optimal ApoE cutoff value for CSF was >1.7 mg/L with 85% sensitivity and 100% specificity and was >42 mg/L in serum with 80% sensitivity and 93% specificity. CONCLUSION ApoE detection provided a novel diagnostic marker for invasive bacterial infections in pediatric patients.
Collapse
Affiliation(s)
- Chuanqing Wang
- The Clinical Microbiology Lab, Department of Nosocomial Infection Control, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | | | | | | | | |
Collapse
|
27
|
Chuang KI, Leung B, Hsu N, Harris HW. Heparin protects against septic mortality via apoE-antagonism. Am J Surg 2011; 202:325-35. [PMID: 21741028 DOI: 10.1016/j.amjsurg.2010.10.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 10/01/2010] [Accepted: 10/01/2010] [Indexed: 10/18/2022]
Abstract
BACKGROUND Apolipoprotein E (apoE), a component of plasma lipoproteins, increases septic mortality in a rodent model of sepsis, presumably by enhancing lipid antigen presentation to antigen-presenting cells via the low-density lipoprotein receptor (LDLR). Downstream, this culminates in natural killer T (NKT) cell activation and cytokine secretion. To determine whether apoE antagonism would protect against septic mortality in mice, apoE-LDLR binding was antagonized using heparin, which can inhibit apoE's LDLR-binding site. METHODS C57BL/6 mice underwent cecal ligation and puncture (CLP) and heparin infusion. Serum partial thromboplastin time and alanine aminotransferase were measured at 24 hours, and survival was monitored for 7 days after CLP. LDLR+/+ and LDLR-/- fibroblasts were incubated with apoE and heparin to measure apoE internalization. Hepatic NKT cells and cytokine levels were quantified via fluorescence-activated cell sorting. RESULTS Heparin decreased CLP-induced mortality by 50% versus saline-treated controls, independent of anticoagulation. LDLR+/+ fibroblasts displayed decreased uptake of apoE when treated concurrently with heparin for 12 hours. In septic mice, hepatic alanine aminotransferase levels, hepatic NKT cells, and plasma cytokine levels decreased after heparin treatment. CONCLUSIONS This study demonstrates that heparin protects against septic mortality independent of its anticoagulant effect. This protective effect is associated with the inhibition of apoE-LDLR binding, diminished NKT proliferation and cytokine production, and hepatic dysfunction. These findings indicate a potential clinical role for apoE antagonism in the treatment of sepsis.
Collapse
Affiliation(s)
- Kelley I Chuang
- Department of Surgery, University of California, San Francisco-East Bay, Oakland, CA, USA
| | | | | | | |
Collapse
|
28
|
Cross-talk between apolipoprotein E and cytokines. Mediators Inflamm 2011; 2011:949072. [PMID: 21772670 PMCID: PMC3136159 DOI: 10.1155/2011/949072] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Accepted: 05/02/2011] [Indexed: 02/06/2023] Open
Abstract
Apolipoprotein E (apoE) is a multifunctional glycosylated protein characterized by its wide tissue distribution. Despite its importance in lipid transport and atherosclerosis pathogenesis, apoE is associated with neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson disease, and autoimmune disorders such as multiple sclerosis and psoriasis. Among others, the role of apoE in modulating inflammation and oxidation is crucial in elucidating the risk factors of the above diseases since the function of apoE is closely linked with both proinflammatory and antiinflammatory cytokines. Moreover, apoE modulates inflammatory and immune responses in an isoform-dependent manner. Correspondingly, inflammatory cytokines can either upregulate or downregulate the production of apoE in various tissue types. However, studies on the interactions between apoE and cytokines occasionally yield conflicting results, highlighting the complex roles of apoE and cytokines in various disorders. The present paper summarizes the current knowledge about the cross-talk between apoE and cytokines, with emphasis on the effects of apoE on the Th1/Th2 balance.
Collapse
|
29
|
Zheng Y, Patel AB, Narayanaswami V, Hura GL, Hang B, Bielicki JK. HDL mimetic peptide ATI-5261 forms an oligomeric assembly in solution that dissociates to monomers upon dilution. Biochemistry 2011; 50:4068-76. [PMID: 21476522 DOI: 10.1021/bi2002955] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
ATI-5261 is a 26-mer peptide that stimulates cellular cholesterol efflux with high potency. This peptide displays high aqueous solubility, despite having amphipathic α-helix structure and a broad nonpolar surface. These features suggested to us that ATI-5261 may adopt a specific form in solution, having favorable structural characteristics and dynamics. To test this, we subjected ATI-5261 to a series of biophysical studies and correlated self-association with secondary structure and activity. Gel-filtration chromatography and native gel electrophoresis indicated ATI-5261 adopted a discrete self-associated form of low molecular weight at concentrations >1 mg/mL. Formation of a discrete molecular species was verified by small-angle X-ray scattering (SAXS), which further revealed the peptide formed a tetrameric assembly having an elongated shape and hollow central core. This assembly dissociated to individual peptide strands upon dilution to concentrations required for promoting high-affinity cholesterol efflux from cells. Moreover, the α-helical content of ATI-5261 was exceptionally high (74.1 ± 6.8%) regardless of physical form and concentration. Collectively, these results indicate ATI-5261 displays oligomeric behavior generally similar to native apolipoproteins and dissociates to monomers of high α-helical content upon dilution. Optimizing self-association behavior and secondary structure may prove useful for improving the translatability and efficacy of apolipoprotein mimetic peptides.
Collapse
Affiliation(s)
- Ying Zheng
- Life Sciences Division, Lawrence Berkeley National Laboratory, Donner Laboratory, University of California, Berkeley, Berkeley, California 94720, United States
| | | | | | | | | | | |
Collapse
|
30
|
Leung B, Harris HW. NKT cells: the culprits of sepsis? J Surg Res 2010; 167:87-95. [PMID: 21035139 DOI: 10.1016/j.jss.2010.09.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 06/08/2010] [Accepted: 09/23/2010] [Indexed: 11/28/2022]
Abstract
Sepsis is currently a leading cause of death in hospital intensive care units. Previous studies suggest that the pathophysiology of sepsis involves the hyperactivation of complex pro-inflammatory cascades that include the activation of various immune cells and the exuberant secretion of pro-inflammatory cytokines by these cells. Natural killer T-cells (NKT) are a sub-lineage of T cells that share characteristics of conventional T cells and NK cells, and bridge innate and adaptive immunity. More recently, NKT cells have been implicated in microbial immunity, including the onset of sepsis. Moreover, apolipoprotein E (apoE), a component of triglyceride-rich lipoproteins, has been shown to be protective in endotoxemia and gram-negative infections in addition to its well-known role in lipid metabolism. Here, we will review the role of NKT cells in sepsis and septic shock, the immunoregulatory role of apoE in the host immune response to infection, and propose a mechanism for this immunoregulation.
Collapse
Affiliation(s)
- Briana Leung
- Department of Surgery, University of California, San Francisco, California 94143-0104, USA
| | | |
Collapse
|
31
|
Abstract
Sepsis is currently a leading cause of death in hospital intensive care units. Previous studies suggest that the pathophysiology of sepsis involves the hyperactivation of complex proinflammatory cascades that include the activation of various immune cells and the exuberant secretion of proinflammatory cytokines by these cells. Natural killer T-cells (NKTs) are a sublineage of T cells that share characteristics of conventional T cells and NK cells and bridge innate and adaptive immunity. More recently, NKT cells have been implicated in microbial immunity, including the onset of sepsis. Moreover, apolipoprotein E (apoE), a component of triglyceride-rich lipoproteins, has been shown to be protective in endotoxemia and gram-negative infections in addition to its well-known role in lipid metabolism. Here, we will review the role of NKT cells in sepsis and septic shock, the immunoregulatory role of apoE in the host immune response to infection, and propose a mechanism for this immunoregulation.
Collapse
|
32
|
Chuang K, Elford EL, Tseng J, Leung B, Harris HW. An expanding role for apolipoprotein E in sepsis and inflammation. Am J Surg 2010; 200:391-7. [PMID: 20409531 PMCID: PMC2909338 DOI: 10.1016/j.amjsurg.2009.10.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 10/19/2009] [Accepted: 10/27/2009] [Indexed: 11/28/2022]
Abstract
BACKGROUND Apolipoprotein E (apoE), a component of plasma lipoproteins, plays an important, but poorly defined role in sepsis. We have shown that injecting apoE increases septic mortality in a rat model of gram-negative bacterial sepsis, with concomitant hepatic natural killer T (NKT) cell proliferation and activation. The presumed mechanism for this apoE-mediated mortality is that apoE can bind and traffic antigens, presumed to include lipopolysaccharide (LPS), and promote activation of dendritic cells (DC) with subsequent NKT activation and cytokine release. Thus, we sought to prove that LPS was the antigen responsible for the increased NKT activation enhanced by the presence of apoE. METHODS We isolated murine marrow-derived DCs, pulsed them with lipid antigen (LPS, and positive controls alpha-galactosylceramide [alpha-GalCer] and isoglobotrihexosylceramide 3 [iGb3]) with or without apoE, and then cocultured the DCs with hybridoma NKTs. NKT activation was measured by interleukin-2 (IL-2) supernatant levels using enzyme-linked immunosorbent assay (ELISA). RESULTS LPS at different concentrations was a weak stimulus for NKT activation regardless of apoE presence. When apoE was present, iGb3, an endogenous ligand analog, elicited more than a 2-fold increase in IL-2 response when compared with iGb3 alone (P < .05). CONCLUSIONS These results indicate an endogenous ligand, not LPS, may be responsible for NKT activation. A molecular remnant similar to iGb3 could act as a damage-associated molecular pattern and play a prominent role in animal models of sepsis.
Collapse
Affiliation(s)
- Kelley Chuang
- Department of Surgery, University of California, San Francisco, East Bay, Oakland, USA
| | | | | | | | | |
Collapse
|
33
|
Hubacek JA, Peasey A, Pikhart H, Stavek P, Kubinova R, Marmot M, Bobak M. APOE polymorphism and its effect on plasma C-reactive protein levels in a large general population sample. Hum Immunol 2010; 71:304-8. [PMID: 20074603 PMCID: PMC2837141 DOI: 10.1016/j.humimm.2010.01.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 12/23/2009] [Accepted: 01/07/2010] [Indexed: 01/26/2023]
Abstract
The published data remain inconsistent on association between apolipoprotein E (APOE) gene variations and plasma levels of C-reactive protein (CRP), mainly because of low statistical power of previous studies. To clarify this question, we analyzed data from large population sample of randomly selected individuals from seven Czech towns (2,886 males and 3,344 females, the HAPIEE [Health, Alcohol, and Psychosocial factors In Eastern Europe] study). In both males and females, the lowest levels of plasma hsCRP were observed in the carriers of the APOE epsilon 4 epsilon 4 and epsilon 4 epsilon 3 genotypes. The median (interquartile range, IQR) concentration of hsCRP in carriers of the most common APOE epsilon 3 epsilon 3 genotype (two-thirds of participants) was 1.13 mg/l (IQR, 0.56-2.33) in men and 1.23 mg/l (IQR, 0.61-2.65) in women, compared with 0.72 mg/l (IQR, 0.61-0.86) in male and 0.72 mg/l (IQR, 0.61-0.85) in female carriers of APOE epsilon 4 epsilon 3/epsilon 4 epsilon 4 genotypes; the differences were statistically significant (p < 0.001). The association between APOE and CRP was not materially affected by adjustment for age, sex, history of cardiovascular disease, or cardiovascular risk factors. This study, the largest to date, provides robust evidence of an association between plasma hsCRP and the APOE genotype, an association not explained by history of cardiovascular disease nor its risk factors.
Collapse
Affiliation(s)
- Jaroslav A. Hubacek
- Institute of Clinical and Experimental Medicine, Prague, Czech Republic
- Centre for Cardiovascular Research, Prague, Czech Republic
- South Bohemia University, Faculty of Public Health and Social Studies, Ceske Budejovice, Czech Republic
| | - Anne Peasey
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Hynek Pikhart
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Petr Stavek
- Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Michael Marmot
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Martin Bobak
- Department of Epidemiology and Public Health, University College London, London, UK
| |
Collapse
|
34
|
The role of apolipoprotein E in Guillain-Barré syndrome and experimental autoimmune neuritis. J Biomed Biotechnol 2010; 2010:357412. [PMID: 20182542 PMCID: PMC2825561 DOI: 10.1155/2010/357412] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 12/20/2009] [Indexed: 11/24/2022] Open
Abstract
Apolipoprotein E (apoE) is a 34.2 kDa glycosylated protein characterized by its wide tissue distribution and multiple functions. ApoE has been widely studied in lipid metabolism, cardiocerebrovascular diseases, and neurodegenerative diseases like Alzheimer's disease and mild cognitive impairment, and so forth. Recently, a growing body of evidence has pointed to nonlipid related properties of apoE, including suppression of T cell proliferation, regulation of macrophage function, facilitation of lipid antigen presentation by CD1 molecules to natural killer T (NKT) cells, and modulation of inflammation and oxidation. By these properties, apoE impacts physiology and pathophysiology at multiple levels. The present paper summarizes updated studies on the immunoregulatory function of apoE, with special focus on isoform-specific effects of apoE on Guillain-Barré syndrome (GBS) and its animal model experimental autoimmune neuritis (EAN).
Collapse
|
35
|
Frazier WJ, Wang X, Wancket LM, Li XA, Meng X, Nelin LD, Cato ACB, Liu Y. Increased inflammation, impaired bacterial clearance, and metabolic disruption after gram-negative sepsis in Mkp-1-deficient mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:7411-9. [PMID: 19890037 DOI: 10.4049/jimmunol.0804343] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
MAPKs are crucial for TNF-alpha and IL-6 production by innate immune cells in response to TLR ligands. MAPK phosphatase 1 (Mkp-1) deactivates p38 and JNK, abrogating the inflammatory response. We have previously demonstrated that Mkp-1(-/-) mice exhibit exacerbated inflammatory cytokine production and increased mortality in response to challenge with LPS and heat-killed Staphylococcus aureus. However, the function of Mkp-1 in host defense during live Gram-negative bacterial infection remains unclear. We challenged Mkp-1(+/+) and Mkp-1(-/-) mice with live Escherichia coli i.v. to examine the effects of Mkp-1 deficiency on animal survival, bacterial clearance, metabolic activity, and cytokine production. We found that Mkp-1 deficiency predisposed animals to accelerated mortality and was associated with more robust production of TNF-alpha, IL-6 and IL-10, greater bacterial burden, altered cyclooxygenase-2 and iNOS expression, and substantial changes in the mobilization of energy stores. Likewise, knockout of Mkp-1 also sensitized mice to sepsis caused by cecal ligation and puncture. IL-10 inhibition by neutralizing Ab or genetic deletion alleviated increased bacterial burden. Treatment with the bactericidal antibiotic gentamicin, given 3 h after Escherichia coli infection, protected Mkp-1(+/+) mice from septic shock but had no effect on Mkp-1(-/-) mice. Thus, during Gram-negative bacterial sepsis Mkp-1 not only plays a critical role in the regulation of cytokine production but also orchestrates the bactericidal activities of the innate immune system and controls the metabolic response to stress.
Collapse
Affiliation(s)
- W Joshua Frazier
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hu CK, Venet F, Heffernan DS, Wang YL, Horner B, Huang X, Chung CS, Gregory SH, Ayala A. The role of hepatic invariant NKT cells in systemic/local inflammation and mortality during polymicrobial septic shock. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:2467-75. [PMID: 19201902 PMCID: PMC2653268 DOI: 10.4049/jimmunol.0801463] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NKT cells have been described as innate regulatory cells because of their rapid response to conserved glycolipids presented on CD1d via their invariant TCR. However, little is known about the contribution of the hepatic NKT cell to the development of a local and/or systemic immune response to acute septic challenge (cecal ligation and puncture (CLP)). We found not only that mice deficient in invariant NKT cells (Jalpha18(-/-)) had a marked attenuation in CLP-induced mortality, but also exhibited an oblation of the systemic inflammatory response (with little effect on splenic/peritoneal immune responsiveness). Flow cytometric data indicated that following CLP, there was a marked decline in the percentage of CD3(+)alpha-galactosylceramide CD1d tetramer(+) cells in the mouse C57BL/6J and BALB/c liver nonparenchymal cell population. This was associated with the marked activation of these cells (increased expression of CD69 and CD25) as well as a rise in the frequency of NKT cells positive for both Th1 and Th2 intracellular cytokines. In this respect, when mice were pretreated in vivo with anti-CD1d-blocking Ab, we observed not only that this inhibited the systemic rise of IL-6 and IL-10 levels in septic mice and improved overall septic survival, but that the CLP-induced changes in liver macrophage IL-6 and IL-10 expressions were inversely effected by this treatment. Together, these findings suggest that the activation of hepatic invariant NKT cells plays a critical role in regulating the innate immune/systemic inflammatory response and survival in a model of acute septic shock.
Collapse
Affiliation(s)
- Caroline K Hu
- Shock-Trauma Research Laboratories, Division of Surgical Research, Department of Surgery, Rhode Island Hospital and Brown University School of Medicine, Providence, RI 02903, USA
| | | | | | | | | | | | | | | | | |
Collapse
|