1
|
Rani A, Stadler JT, Marsche G. HDL-based therapeutics: A promising frontier in combating viral and bacterial infections. Pharmacol Ther 2024; 260:108684. [PMID: 38964560 DOI: 10.1016/j.pharmthera.2024.108684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Low levels of high-density lipoprotein (HDL) and impaired HDL functionality have been consistently associated with increased susceptibility to infection and its serious consequences. This has been attributed to the critical role of HDL in maintaining cellular lipid homeostasis, which is essential for the proper functioning of immune and structural cells. HDL, a multifunctional particle, exerts pleiotropic effects in host defense against pathogens. It functions as a natural nanoparticle, capable of sequestering and neutralizing potentially harmful substances like bacterial lipopolysaccharides. HDL possesses antiviral activity, preventing viruses from entering or fusing with host cells, thereby halting their replication cycle. Understanding the complex relationship between HDL and the immune system may reveal innovative targets for developing new treatments to combat infectious diseases and improve patient outcomes. This review aims to emphasize the role of HDL in influencing the course of bacterial and viral infections and its and its therapeutic potential.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Styria, Austria.
| |
Collapse
|
2
|
Zhao D, Han X, Mu Q, Wu Y, Shan L, Su L, Wang W, Wang P, Kang Y, Wang F. Association of cerebrospinal fluid NPY with peripheral ApoA: a moderation effect of BMI. Nutr Metab (Lond) 2024; 21:52. [PMID: 39054540 PMCID: PMC11270855 DOI: 10.1186/s12986-024-00828-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Apoprotein A-I (ApoA-I) and Apoprotein B (ApoB) have emerged as novel cardiovascular risk biomarkers influenced by feeding behavior. Hypothalamic appetite peptides regulate feeding behavior and impact lipoprotein levels, which effects vary in different weight states. This study explores the intricate relationship between body mass index (BMI), hypothalamic appetite peptides, and apolipoproteins with emphasis on the moderating role of body weight in the association between neuropeptide Y (NPY), ghrelin, orexin A (OXA), oxytocin in cerebrospinal fluid (CSF) and peripheral ApoA-I and ApoB. METHODS In this cross-sectional study, we included participants with a mean age of 31.77 ± 10.25 years, categorized into a normal weight (NW) (n = 73) and an overweight/obese (OW/OB) (n = 117) group based on BMI. NPY, ghrelin, OXA, and oxytocin levels in CSF were measured. RESULTS In the NW group, peripheral ApoA-I levels were higher, while ApoB levels were lower than in the OW/OB group (all p < 0.05). CSF NPY exhibited a positive correlation with peripheral ApoA-I in the NW group (r = 0.39, p = 0.001). Notably, participants with higher CSF NPY levels had higher peripheral ApoA-I levels in the NW group and lower peripheral ApoA-I levels in the OW/OB group, showing the significant moderating effect of BMI on this association (R2 = 0.144, β=-0.54, p < 0.001). The correlation between ghrelin, OXA and oxytocin in CSF and peripheral ApoB in both groups exhibited opposing trends (Ghrelin: r = -0.03 and r = 0.04; OXA: r = 0.23 and r=-0.01; Oxytocin: r=-0.09 and r = 0.04). CONCLUSION This study provides hitherto undocumented evidence that BMI moderates the relationship between CSF NPY and peripheral ApoA-I levels. It also reveals the protective role of NPY in the NW population, contrasting with its risk factor role in the OW/OB population, which was associated with the at-risk for cardiovascular disease.
Collapse
Affiliation(s)
- Danyang Zhao
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Xiaoli Han
- Clinical Nutrition Department, Friendship hospital of Urumqi in Xinjiang, Urumqi, 830049, China
| | - Qingshuang Mu
- Xinjiang Key Laboratory of Neurological Disorder Research, the Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China
| | - Yan Wu
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China
| | - Ligang Shan
- Department of Anesthesiology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen, 361021, China
| | - Lidong Su
- Department of Anesthesiology, the Third Affiliated Hospital of Inner Mongolia Medical University, BaoGang Hospital, Baotou, 014010, China
| | - Wenyan Wang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Pengxiang Wang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Yimin Kang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, 010110, China.
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China.
| |
Collapse
|
3
|
Mousa H, Al Saei A, Razali RM, Zughaier SM. Vitamin D status affects proteomic profile of HDL-associated proteins and inflammatory mediators in dyslipidemia. J Nutr Biochem 2024; 123:109472. [PMID: 37863441 DOI: 10.1016/j.jnutbio.2023.109472] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023]
Abstract
Vitamin D deficiency and dyslipidemia have substantial implications for human health globally. Vitamin D is essential for bone metabolism and immune modulation, and its insufficiency is linked to various chronic inflammatory conditions. Dyslipidemia, characterized by low levels of high-density lipoprotein (HDL) and elevated levels of low-density lipoprotein (LDL) and triglycerides, is also prevalent. Previous research has shown a connection between vitamin D deficiency and low HDL, but the precise mechanism by which vitamin D influences HDL production and its anti-inflammatory properties remains unclear. This study aimed to investigate the proteomic profiles of individuals with and without vitamin D deficiency and dyslipidemia, specifically focusing on the effects of vitamin D on HDL production, its anti-inflammatory potential, and the molecular pathways associated with vitamin D deficiency and dyslipidemia, particularly inflammation and cancer pathways. By analyzing the proteomic profiles of 274 participants from the Qatar Biobank database, we identified 1301 proteins. Our findings indicated a decrease in HDL-associated apolipoproteins (ApoM and ApoD) in individuals with both dyslipidemia and vitamin D deficiency. Conversely, participants with these conditions exhibited increased expression of acute-phase proteins (SAA1 and SOD1), which are associated with inflammation. Pathway enrichment analysis revealed heightened inflammatory activity in individuals with vitamin D deficiency and dyslipidemia, with notable enrichments in pathways such as MAPK, JAK-STAT, Ras signaling, cytokine-cytokine receptor interaction, AGE-RAGE, ErbB signaling, and cancer pathways. Overall, cases of vitamin D deficiency showed enrichment in inflammation pathways, while individuals with both vitamin D deficiency and dyslipidemia demonstrated enhanced activation of cancer and inflammation pathways.
Collapse
Affiliation(s)
- Hanaa Mousa
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Aisha Al Saei
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Rozaimi Mohamad Razali
- Department of Biomedical Sciences College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Susu M Zughaier
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
4
|
Alkudmani ZS, Alshammary AF, Ali Khan I. Molecular Effect of Variants in Toll-like Receptor 4 Gene in Saudi Patients with Type 2 Diabetes Mellitus. Cells 2023; 12:2340. [PMID: 37830554 PMCID: PMC10571932 DOI: 10.3390/cells12192340] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Single-nucleotide polymorphisms (SNPs) in the Toll-like receptor 4 (TLR4) gene have been documented in type 2 diabetes mellitus (T2DM) and other diseases in the Saudi population. We investigated the relationship between rs11536889, rs4986790, and rs4986791 SNPs in the TLR4 gene and T2DM in the Saudi population; 105 patients with T2DM and 105 healthy controls were analyzed. The TLR4 gene was amplified through PCR, followed by restriction fragment length polymorphism analysis for rs4986791 and Sanger sequencing for rs11536889 and rs4986790 SNPs. The clinical and biochemical characteristics were associated with T2DM (p < 0.05). The rs11536889, rs4986790, and rs4986791 SNPs in control subjects followed the Hardy-Weinberg equilibrium (p > 0.05). Alleles were associated with rs11536889, rs4986791, heterozygous codominant, and dominant models (p < 0.05). However, the rs4986790 SNP was not associated with T2DM (p > 0.05). Logistic regression analysis showed that high-density lipoprotein cholesterol (HDLc) levels were associated with T2DM (p < 0.001). Analysis of variance showed that waist (p = 0.0005) and hip circumferences (p = 0.002) in rs4986790 and rs4986791 SNPs, in SBP (p = 0.001), DBP (p = 0.002), and HDLc levels (p = 0.003), were associated with T2DM subjects. T2DM was also associated with the haplotype (p < 0.001) but not with linkage disequilibrium. The gene-gene interaction was associated with the three SNPs studied in patients with T2DM according to the generalized multifactor dimensionality reduction model (p < 0.0001). Dendrogram and graphical depletion analysis revealed a moderate association in patients with T2DM. The results suggest that rs11536889 and rs4986790 SNPs are genotypically and allelically associated with T2DM in Saudi patients. Future functional studies are recommended to validate the genetic roles of these SNPs in the pathogenesis and progression of diseases.
Collapse
Affiliation(s)
| | | | - Imran Ali Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (Z.S.A.); (A.F.A.)
| |
Collapse
|
5
|
Rani A, Marsche G. A Current Update on the Role of HDL-Based Nanomedicine in Targeting Macrophages in Cardiovascular Disease. Pharmaceutics 2023; 15:1504. [PMID: 37242746 PMCID: PMC10221824 DOI: 10.3390/pharmaceutics15051504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
High-density lipoproteins (HDL) are complex endogenous nanoparticles involved in important functions such as reverse cholesterol transport and immunomodulatory activities, ensuring metabolic homeostasis and vascular health. The ability of HDL to interact with a plethora of immune cells and structural cells places it in the center of numerous disease pathophysiologies. However, inflammatory dysregulation can lead to pathogenic remodeling and post-translational modification of HDL, rendering HDL dysfunctional or even pro-inflammatory. Monocytes and macrophages play a critical role in mediating vascular inflammation, such as in coronary artery disease (CAD). The fact that HDL nanoparticles have potent anti-inflammatory effects on mononuclear phagocytes has opened new avenues for the development of nanotherapeutics to restore vascular integrity. HDL infusion therapies are being developed to improve the physiological functions of HDL and to quantitatively restore or increase the native HDL pool. The components and design of HDL-based nanoparticles have evolved significantly since their initial introduction with highly anticipated results in an ongoing phase III clinical trial in subjects with acute coronary syndrome. The understanding of mechanisms involved in HDL-based synthetic nanotherapeutics is critical to their design, therapeutic potential and effectiveness. In this review, we provide a current update on HDL-ApoA-I mimetic nanotherapeutics, highlighting the scope of treating vascular diseases by targeting monocytes and macrophages.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
6
|
Kotlyarov S. High-Density Lipoproteins: A Role in Inflammation in COPD. Int J Mol Sci 2022; 23:8128. [PMID: 35897703 PMCID: PMC9331387 DOI: 10.3390/ijms23158128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a widespread disease associated with high rates of disability and mortality. COPD is characterized by chronic inflammation in the bronchi as well as systemic inflammation, which contributes significantly to the clinically heterogeneous course of the disease. Lipid metabolism disorders are common in COPD, being a part of its pathogenesis. High-density lipoproteins (HDLs) are not only involved in lipid metabolism, but are also part of the organism's immune and antioxidant defense. In addition, HDL is a versatile transport system for endogenous regulatory agents and is also involved in the removal of exogenous substances such as lipopolysaccharide. These functions, as well as information about lipoprotein metabolism disorders in COPD, allow a broader assessment of their role in the pathogenesis of heterogeneous and comorbid course of the disease.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
7
|
Stasi A, Franzin R, Fiorentino M, Squiccimarro E, Castellano G, Gesualdo L. Multifaced Roles of HDL in Sepsis and SARS-CoV-2 Infection: Renal Implications. Int J Mol Sci 2021; 22:5980. [PMID: 34205975 PMCID: PMC8197836 DOI: 10.3390/ijms22115980] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023] Open
Abstract
High-density lipoproteins (HDLs) are a class of blood particles, principally involved in mediating reverse cholesterol transport from peripheral tissue to liver. Omics approaches have identified crucial mediators in the HDL proteomic and lipidomic profile, which are involved in distinct pleiotropic functions. Besides their role as cholesterol transporter, HDLs display anti-inflammatory, anti-apoptotic, anti-thrombotic, and anti-infection properties. Experimental and clinical studies have unveiled significant changes in both HDL serum amount and composition that lead to dysregulated host immune response and endothelial dysfunction in the course of sepsis. Most SARS-Coronavirus-2-infected patients admitted to the intensive care unit showed common features of sepsis disease, such as the overwhelmed systemic inflammatory response and the alterations in serum lipid profile. Despite relevant advances, episodes of mild to moderate acute kidney injury (AKI), occurring during systemic inflammatory diseases, are associated with long-term complications, and high risk of mortality. The multi-faceted relationship of kidney dysfunction with dyslipidemia and inflammation encourages to deepen the clarification of the mechanisms connecting these elements. This review analyzes the multifaced roles of HDL in inflammatory diseases, the renal involvement in lipid metabolism, and the novel potential HDL-based therapies.
Collapse
Affiliation(s)
- Alessandra Stasi
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (M.F.)
| | - Rossana Franzin
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (M.F.)
| | - Marco Fiorentino
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (M.F.)
| | - Enrico Squiccimarro
- Department of Emergency and Organ Transplant (DETO), University of Bari, 70124 Bari, Italy;
- Cardio-Thoracic Surgery Department, Heart & Vascular Centre, Maastricht University Medical Centre (MUMC), 6229HX Maastricht, The Netherlands
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy;
| | - Loreto Gesualdo
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (M.F.)
| |
Collapse
|
8
|
Trakaki A, Marsche G. Current Understanding of the Immunomodulatory Activities of High-Density Lipoproteins. Biomedicines 2021; 9:biomedicines9060587. [PMID: 34064071 PMCID: PMC8224331 DOI: 10.3390/biomedicines9060587] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Lipoproteins interact with immune cells, macrophages and endothelial cells - key players of the innate and adaptive immune system. High-density lipoprotein (HDL) particles seem to have evolved as part of the innate immune system since certain HDL subspecies contain combinations of apolipoproteins with immune regulatory functions. HDL is enriched in anti-inflammatory lipids, such as sphingosine-1-phosphate and certain saturated lysophospholipids. HDL reduces inflammation and protects against infection by modulating immune cell function, vasodilation and endothelial barrier function. HDL suppresses immune cell activation at least in part by modulating the cholesterol content in cholesterol/sphingolipid-rich membrane domains (lipid rafts), which play a critical role in the compartmentalization of signaling pathways. Acute infections, inflammation or autoimmune diseases lower HDL cholesterol levels and significantly alter HDL metabolism, composition and function. Such alterations could have a major impact on disease progression and may affect the risk for infections and cardiovascular disease. This review article aims to provide a comprehensive overview of the immune cell modulatory activities of HDL. We focus on newly discovered activities of HDL-associated apolipoproteins, enzymes, lipids, and HDL mimetic peptides.
Collapse
|
9
|
Oehler B, Kloka J, Mohammadi M, Ben-Kraiem A, Rittner HL. D-4F, an ApoA-I mimetic peptide ameliorating TRPA1-mediated nocifensive behaviour in a model of neurogenic inflammation. Mol Pain 2020; 16:1744806920903848. [PMID: 31996074 PMCID: PMC6993174 DOI: 10.1177/1744806920903848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background High doses of capsaicin are recommended for the treatment of neuropathic pain. However, low doses evoke mechanical hypersensitivity. Activation of the capsaicin chemosensor transient receptor potential vanilloid 1 (TRPV1) induces neurogenic inflammation. In addition to the release of pro-inflammatory mediators, reactive oxygen species are produced. These highly reactive molecules generate oxidised phospholipids and 4-hydroxynonenal (4-HNE) which then directly activate TRP ankyrin 1 (TRPA1). The apolipoprotein A-I mimetic peptide D-4F neutralises oxidised phospholipids. Here, we asked whether D-4F ameliorates neurogenic hypersensitivity in rodents by targeting reactive oxygen species and 4-HNE in the capsaicin-evoked pain model. Results Co-application of D-4F ameliorated capsaicin-induced mechanical hypersensitivity and allodynia as well as persistent heat hypersensitivity measured by Randell–Selitto, von Frey and Hargreaves test, respectively. In addition, mechanical hypersensitivity was blocked after co-injection of D-4F with the reactive oxygen species analogue H2O2 or 4-HNE. In vitro studies on dorsal root ganglion neurons and stably transfected cell lines revealed a TRPA1-dependent inhibition of the calcium influx when agonists were pre-incubated with D-4F. The capsaicin-induced calcium influx in TRPV1-expressing cell lines and dorsal root ganglion neurons sustained in the presence of D-4F. Conclusions D-4F is a promising compound to ameliorate TRPA1-dependent hypersensitivity during neurogenic inflammation.
Collapse
Affiliation(s)
- Beatrice Oehler
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany.,Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Jan Kloka
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany.,Department of Anaesthesiology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Milad Mohammadi
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany.,Department of Anaesthesiology, University Hospital of Cologne, Cologne, Germany
| | - Adel Ben-Kraiem
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany
| | - Heike L Rittner
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
10
|
Amiri P, Naghizadeh M, Baradaran B, Saghafi-Asl M, Shanehbandi D, Mirmajidi S. Insulin resistance in relation to inflammatory gene expression and metabolic features in apparently healthy obese individuals. Int J Diabetes Dev Ctries 2019. [DOI: 10.1007/s13410-018-0626-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
11
|
Chen X, Tang L, Feng J, Wang Y, Han Z, Meng J. Downregulation of Paralemmin-3 Ameliorates Lipopolysaccharide-Induced Acute Lung Injury in Rats by Regulating Inflammatory Response and Inhibiting Formation of TLR4/MyD88 and TLR4/TRIF Complexes. Inflammation 2018; 40:1983-1999. [PMID: 28801798 PMCID: PMC7102376 DOI: 10.1007/s10753-017-0639-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have demonstrated paralemmin-3 (PALM3) participates in Toll-like receptor (TLR) signaling. This study investigated the effect of PALM3 knockdown on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and its underlying mechanisms. We constructed a recombinant adenoviral vector containing short hairpin RNA for PALM3 to knockdown PALM3 expression. A transgene-free adenoviral vector was used as a negative control. The ALI rat model was established by LPS peritoneal injection at 48-h post-transfection. Results showed that downregulation of PALM3 improved the survival rate, attenuated lung pathological changes, alleviated pulmonary edema, lung vascular leakage and neutrophil infiltration, inhibited the production of proinflammatory cytokines and activation of nuclear factor κB and interferon β regulatory factor 3, and promoted the secretion of anti-inflammatory cytokine interleukin-10 and expression of suppressor of cytokine signaling-3 in the ALI rat model. However, PALM3 knockdown had no effect on TLR4, myeloid differentiation factor 88 (MyD88), and Toll-interleukin-1 receptor domain-containing adaptor inducing interferon β (TRIF) expression. Moreover, PALM3 knockdown reduced the interaction of TLR4 with MyD88 or TRIF induced by LPS in rat lungs. Therefore, the downregulation of PALM3 protected rats from LPS-induced ALI and its mechanisms were partially associated with the modulation of inflammatory responses and inhibition of TLR4/MyD88 and TLR4/TRIF complex formation.
Collapse
Affiliation(s)
- Xuxin Chen
- Department of Respiratory Medicine, Navy General Hospital of the PLA, No. 6 Fucheng Road, Beijing, 100037, China
| | - Lu Tang
- Department of Neurology, The First Hospital of Changsha, Changsha, 430100, People's Republic of China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yi Wang
- Department of Respiratory Medicine, The Sixth People's Hospital of Jinan City Affiliated to Jining Medical College, Jinan, 250200, People's Republic of China
| | - Zhihai Han
- Department of Respiratory Medicine, Navy General Hospital of the PLA, No. 6 Fucheng Road, Beijing, 100037, China.
| | - Jiguang Meng
- Department of Respiratory Medicine, Navy General Hospital of the PLA, No. 6 Fucheng Road, Beijing, 100037, China.
| |
Collapse
|
12
|
Kareinen I, Baumann M, Nguyen SD, Maaninka K, Anisimov A, Tozuka M, Jauhiainen M, Lee-Rueckert M, Kovanen PT. Chymase released from hypoxia-activated cardiac mast cells cleaves human apoA-I at Tyr 192 and compromises its cardioprotective activity. J Lipid Res 2018; 59:945-957. [PMID: 29581158 DOI: 10.1194/jlr.m077503] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 03/22/2018] [Indexed: 01/05/2023] Open
Abstract
ApoA-I, the main structural and functional protein of HDL particles, is cardioprotective, but also highly sensitive to proteolytic cleavage. Here, we investigated the effect of cardiac mast cell activation and ensuing chymase secretion on apoA-I degradation using isolated rat hearts in the Langendorff perfusion system. Cardiac mast cells were activated by injection of compound 48/80 into the coronary circulation or by low-flow myocardial ischemia, after which lipid-free apoA-I was injected and collected in the coronary effluent for cleavage analysis. Mast cell activation by 48/80 resulted in apoA-I cleavage at sites Tyr192 and Phe229, but hypoxic activation at Tyr192 only. In vitro, the proteolytic end-product of apoA-I with either rat or human chymase was the Tyr192-truncated fragment. This fragment, when compared with intact apoA-I, showed reduced ability to promote migration of cultured human coronary artery endothelial cells in a wound-healing assay. We propose that C-terminal truncation of apoA-I by chymase released from cardiac mast cells during ischemia impairs the ability of apoA-I to heal damaged endothelium in the ischemic myocardium.
Collapse
Affiliation(s)
- Ilona Kareinen
- Wihuri Research Institute, Helsinki, Finland; Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Marc Baumann
- Protein Chemistry Unit, Institute of Biomedicine/Anatomy, University of Helsinki, Helsinki, Finland
| | | | | | - Andrey Anisimov
- Wihuri Research Institute, Helsinki, Finland; Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Minoru Tozuka
- Analytical Laboratory Chemistry, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; National Institute for Health and Welfare, Helsinki, Finland
| | | | | |
Collapse
|
13
|
Tschöpe C, Van Linthout S, Kherad B. Heart Failure with Preserved Ejection Fraction and Future Pharmacological Strategies: a Glance in the Crystal Ball. Curr Cardiol Rep 2017; 19:70. [PMID: 28656481 DOI: 10.1007/s11886-017-0874-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The current definition of heart failure is mainly based on an inappropriate measure of cardiac function, i.e., left ventricular ejection fraction (LVEF). The initial sole entity, heart failure with reduced ejection fraction (HFrEF, LVEF <40%), was complemented by the addition of heart failure with preserved ejection fraction (HFpEF, LVEF ≥50%) and most recently, heart failure with mid-range ejection fraction (HFmrEF, LVEF 40-49%). Initially, HFpEF was believed to be a purely left ventricular diastolic dysfunction. Pathophysiological concepts of HFpEF have changed considerably during the last years. In addition to intrinsic cardiac mechanisms, the heart failure pathogenesis is increasingly considered as driven by non-cardiac systemic processes including metabolic disorders, ischemic conditions, and pro-inflammatory/pro-fibrotic or immunological alterations. Presentation and pathophysiology of HFpEF is heterogeneous, and its management remains a challenge since evidence of therapeutic benefits is scarce. Up to now, there are no therapies improving survival in patients with HFpEF. RECENT FINDINGS Several results from clinical and preclinical interventions targeting non-cardiac mechanisms or non-pharmacological interventions including new anti-diabetic or anti-inflammatory drugs, mitochondrial-targeted anti-oxidants, anti-fibrotic strategies, microRNases incl. antagomirs, cell therapeutic options, and high-density lipoprotein-raising strategies are promising and under further investigation. This review addresses mechanisms and available data of current best clinical practice and novel approaches towards HFpEF.
Collapse
Affiliation(s)
- Carsten Tschöpe
- Department of Cardiology, Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany. .,Berliner Zentrum für Regenerative Therapien (BCRT), Campus Virchow Klinikum (CVK), Berlin, Germany. .,Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Berlin, Germany. .,Campus Virchow Clinic, Department of Cardiology, Charité - Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany.
| | - Sophie Van Linthout
- Berliner Zentrum für Regenerative Therapien (BCRT), Campus Virchow Klinikum (CVK), Berlin, Germany.,Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Berlin, Germany.,Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Föhrerstrasse 15, 13353, Berlin, Germany
| | - Behrouz Kherad
- Department of Cardiology, Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,Campus Virchow Clinic, Department of Cardiology, Charité - Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany.,Privatpraxis Dr. Kherad, Große Hamburger Strasse 5-11, 10115, Berlin, Germany
| |
Collapse
|
14
|
Chen XX, Tang L, Fu YM, Wang Y, Han ZH, Meng JG. Paralemmin-3 contributes to lipopolysaccharide-induced inflammatory response and is involved in lipopolysaccharide-Toll-like receptor-4 signaling in alveolar macrophages. Int J Mol Med 2017; 40:1921-1931. [PMID: 29039447 DOI: 10.3892/ijmm.2017.3161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/19/2017] [Indexed: 11/06/2022] Open
Abstract
Alveolar macrophages (AMs) are the first line of defense against foreign stimulation in alveoli, and they participate in inflammatory responses during acute lung injury (ALI). Previous studies indicated that paralemmin-3 (PALM3) expression is induced by lipopolysaccharides (LPS) and may be involved in LPS-Toll-like receptor 4 (TLR4) signaling in alveolar epithelial cells. The aim of the present study was to investigate the effect of PALM3 on LPS-induced inflammation and its underlying mechanisms in rat AMs. For this purpose, the authors detected the expression of PALM3 in AMs by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting following LPS stimulation. Following this, a recombinant adenovirus expressing short hairpin RNA (shRNA) for PALM3 was constructed, as well as a recombinant adenovirus carrying the rat PALM3 gene to modulate the expression of PALM3 in rat AMs. At 48 h after transfection, the PALM3 expression in AMs was detected by RT-qPCR and western blotting. The levels of several cytokines and the activity of nuclear factor-κB and interferon regulatory factor 3 in AMs were measured after LPS stimulation. The localization of PALM3 and LPS-TLR4 signaling adaptor molecules in AMs was analyzed by confocal microscopy, and the physical interactions of PALM3 with these adaptors were assessed by co-immunoprecipitation assays. LPS induced PALM3 expression in AMs and that PALM3 expression promoted the LPS-induced inflammatory response, while PALM3 downregulation suppressed the LPS-induced inflammatory response in AMs. In addition, the results demonstrated that PALM3 could interact with TLR4, myeloid differentiation factor 88, interleukin (IL)-1 receptor associated kinase-1, tumor necrosis factor receptor associated factor-6, and Toll-IL-1 receptor containing adapter molecule-2 in AMs after LPS stimulation. These results suggested that PALM3 contributes to the LPS-induced inflammatory response and participates in LPS-TLR4 signaling in AMs. These data may provide the basis for the development of novel targeted therapeutic strategies of treating ALI.
Collapse
Affiliation(s)
- Xu-Xin Chen
- Department of Respiratory Medicine, Navy General Hospital of the PLA, Beijing 100037, P.R. China
| | - Lu Tang
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan 430100, P.R. China
| | - Yu-Mei Fu
- Department of Emergency, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Yi Wang
- Department of Respiratory Medicine, The Sixth People's Hospital of Jinan City Affiliated to Jining Medical College, Jinan, Shandong 250200, P.R. China
| | - Zhi-Hai Han
- Department of Respiratory Medicine, Navy General Hospital of the PLA, Beijing 100037, P.R. China
| | - Ji-Guang Meng
- Department of Respiratory Medicine, Navy General Hospital of the PLA, Beijing 100037, P.R. China
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW With the intention to summarize the currently available evidence on the pathophysiological relevance of inflammation in heart failure, this review addresses the question whether inflammation is a cause or consequence of heart failure, or both. RECENT FINDINGS This review discusses the diversity (sterile, para-inflammation, chronic inflammation) and sources of inflammation and gives an overview of how inflammation (local versus systemic) can trigger heart failure. On the other hand, the review is outlined how heart failure-associated wall stress and signals released by stressed, malfunctioning, or dead cells (DAMPs: e.g., mitochondrial DNA, ATP, S100A8, matricellular proteins) induce cardiac sterile inflammation and how heart failure provokes inflammation in various peripheral tissues in a direct (inflammatory) and indirect (hemodynamic) manner. The crosstalk between the heart and peripheral organs (bone marrow, spleen, gut, adipose tissue) is outlined and the importance of neurohormonal mechanisms including the renin angiotensin aldosteron system and the ß-adrenergic nervous system in inflammation and heart failure is discussed. Inflammation and heart failure are strongly interconnected and mutually reinforce each other. This indicates the difficulty to counteract inflammation and heart failure once this chronic vicious circle has started and points out the need to control the inflammatory process at an early stage avoiding chronic inflammation and heart failure. The diversity of inflammation further addresses the need for a tailored characterization of inflammation enabling differentiation of inflammation and subsequent target-specific strategies. It is expected that the characterization of the systemic and/or cardiac immune profile will be part of precision medicine in the future of cardiology.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Tschöpe
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Cardiology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
16
|
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
|
17
|
Gordon EM, Figueroa DM, Barochia AV, Yao X, Levine SJ. High-density Lipoproteins and Apolipoprotein A-I: Potential New Players in the Prevention and Treatment of Lung Disease. Front Pharmacol 2016; 7:323. [PMID: 27708582 PMCID: PMC5030281 DOI: 10.3389/fphar.2016.00323] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/05/2016] [Indexed: 12/18/2022] Open
Abstract
Apolipoprotein A-I (apoA-I) and high-density lipoproteins (HDL) mediate reverse cholesterol transport out of cells. Furthermore, HDL has additional protective functions, which include anti-oxidative, anti-inflammatory, anti-apoptotic, and vasoprotective effects. In contrast, HDL can become dysfunctional with a reduction in both cholesterol efflux and anti-inflammatory properties in the setting of disease or the acute phase response. These paradigms are increasingly being recognized to be active in the pulmonary system, where apoA-I and HDL have protective effects in normal lung health, as well as in a variety of disease states, including acute lung injury (ALI), asthma, chronic obstructive pulmonary disease, lung cancer, pulmonary arterial hypertension, pulmonary fibrosis, and viral pneumonia. Similar to observations in cardiovascular disease, however, HDL may become dysfunctional and contribute to disease pathogenesis in respiratory disorders. Furthermore, synthetic apoA-I mimetic peptides have been shown to have protective effects in animal models of ALI, asthma, pulmonary hypertension, and influenza pneumonia. These findings provide evidence to support the concept that apoA-I mimetic peptides might be developed into a new treatment that can either prevent or attenuate the manifestations of lung diseases, such as asthma. Thus, the lung is positioned to take a page from the cardiovascular disease playbook and utilize the protective properties of HDL and apoA-I as a novel therapeutic approach.
Collapse
Affiliation(s)
- Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Debbie M Figueroa
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
18
|
Spillmann F, De Geest B, Muthuramu I, Amin R, Miteva K, Pieske B, Tschöpe C, Van Linthout S. Apolipoprotein A-I gene transfer exerts immunomodulatory effects and reduces vascular inflammation and fibrosis in ob/ob mice. JOURNAL OF INFLAMMATION-LONDON 2016; 13:25. [PMID: 27486384 PMCID: PMC4969975 DOI: 10.1186/s12950-016-0131-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/19/2016] [Indexed: 12/31/2022]
Abstract
Background Obesity is associated with vascular inflammation, fibrosis and reduced high-density lipoproteins (HDL)-cholesterol. We aimed to investigate whether adenoviral gene transfer with human apolipoprotein (apo) A-I (Ad.A-I), the main apo of HDL, could exert immunomodulatory effects and counteract vascular inflammation and fibrosis in ob/ob mice. Methods Ad.A-I transfer was performed in 8 weeks (w) old ob/ob mice, which were sacrificed 7 w later. The aorta was excised for mRNA analysis and the spleen for splenocyte isolation for subsequent flow cytometry and co-culture with murine fibroblasts. HDL was added to mononuclear cells (MNC) and fibroblasts to assess their impact on adhesion capacity and collagen deposition, respectively. Results Ad.A-I led to a 1.8-fold (p < 0.05) increase in HDL-cholesterol versus control ob/ob mice at the day of sacrifice, which was paralleled by a decrease in aortic TNF-α and VCAM-1 mRNA expression. Pre-culture of MNC with HDL decreased their adhesion to TNF-α-activated HAEC. Ad.A-I exerted immunomodulatory effects as evidenced by a downregulation of aortic NOD2 and NLRP3 mRNA expression and by a 12 %, 6.9 %, and 15 % decrease of the induced proliferation/activity of total splenic MNC, CD4+, and CD8+ cells in ob/ob Ad.A-I versus control ob/ob mice, respectively (p < 0.05). Ad.A-I further reduced aortic collagen I and III mRNA expression by 62 % and 66 %, respectively (p < 0.0005), and abrogated the potential of ob/ob splenocytes to induce the collagen content in murine fibroblasts upon co-culture. Finally, HDL decreased the TGF-ß1-induced collagen deposition of murine fibroblasts in vitro. Conclusions Apo A-I transfer counteracts vascular inflammation and fibrosis in ob/ob mice. Electronic supplementary material The online version of this article (doi:10.1186/s12950-016-0131-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Frank Spillmann
- Department of Cardiology, Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Bart De Geest
- Catholic University of Leuven, Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Leuven, Belgium
| | - Ilayaraja Muthuramu
- Catholic University of Leuven, Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Leuven, Belgium
| | - Ruhul Amin
- Catholic University of Leuven, Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Leuven, Belgium
| | - Kapka Miteva
- Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Südstrasse 2, 13353 Berlin, Germany
| | - Burkert Pieske
- Department of Cardiology, Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany ; Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Berlin, Germany ; Department of Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology, Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany ; Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Südstrasse 2, 13353 Berlin, Germany ; Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Berlin, Germany
| | - Sophie Van Linthout
- Department of Cardiology, Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany ; Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Südstrasse 2, 13353 Berlin, Germany ; Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Berlin, Germany
| |
Collapse
|
19
|
Zou G, He J, Ren B, Xu F, Xu G, Zhang W. The delta high-density lipoprotein cholesterol ratio: a novel parameter for gram-negative sepsis. SPRINGERPLUS 2016; 5:1044. [PMID: 27462492 PMCID: PMC4940328 DOI: 10.1186/s40064-016-2685-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 06/23/2016] [Indexed: 01/18/2023]
Abstract
Objective To study changes in blood lipid metabolism in sepsis patients, especially high-density lipoprotein cholesterol (HDL-C) changes in the diagnosis of sepsis and the type of bacteria involved. Methods Two-hundred-twenty cases of patients with febrile infections were divided into local infection, systemic inflammatory response syndrome or sepsis (sepsis) group. For controls, 81 cases of patients with a healthy check-up were used. Lipid levels and inflammatory state were supervised, and a comparative analysis of patients admitted to the hospital after 1, 5, 10 days was performed. Results In patients with sepsis, total cholesterol, HDL-C, and apolipoprotein A 1 (apoA 1) were significantly decreased in this group. Particularly HDL-C was decreased 1 day after admission. Compared with the patients with gram-positive sepsis, HDL-C and apoA1 were significantly reduced in the patients with gram-negative sepsis at admission. The 24-h change ratio of HDL-C was different between the gram-negative and gram-positive sepsis patients with a 70.5 % specificity and 76.5 % sensitivity. The area under the curve was 0.744, and the critical value was −21.1 %. Conclusions The sepsis patients had lower HDL-C than the other groups. The 24-h change ratio of HDL-C can be used as a sepsis diagnosis maker and to distinguish between the bacteria involved in sepsis.
Collapse
Affiliation(s)
- Guoying Zou
- Department of Clinical Laboratory, Brain Hospital of Hunan Province, Furong Middle Road 427, Changsha, 410007 Hunan People's Republic of China ; Department of Medical Laboratory, Hunan University of Traditional Chinese Medicine, Changsha, People's Republic of China
| | - Junyu He
- Department of Clinical Laboratory, Brain Hospital of Hunan Province, Furong Middle Road 427, Changsha, 410007 Hunan People's Republic of China ; Department of Medical Laboratory, Hunan University of Traditional Chinese Medicine, Changsha, People's Republic of China
| | - Biqiong Ren
- Department of Clinical Laboratory, Brain Hospital of Hunan Province, Furong Middle Road 427, Changsha, 410007 Hunan People's Republic of China ; Department of Medical Laboratory, Hunan University of Traditional Chinese Medicine, Changsha, People's Republic of China
| | - Fei Xu
- Department of Clinical Laboratory, Brain Hospital of Hunan Province, Furong Middle Road 427, Changsha, 410007 Hunan People's Republic of China ; Department of Medical Laboratory, Hunan University of Traditional Chinese Medicine, Changsha, People's Republic of China
| | - Guofeng Xu
- Department of Clinical Laboratory, Brain Hospital of Hunan Province, Furong Middle Road 427, Changsha, 410007 Hunan People's Republic of China ; Department of Medical Laboratory, Hunan University of Traditional Chinese Medicine, Changsha, People's Republic of China
| | - Wenling Zhang
- Department of Laboratory Medcine, Xiangya Medical School, Central South University, Changsha, People's Republic of China
| |
Collapse
|
20
|
Sultana A, Cochran BJ, Tabet F, Patel M, Torres LC, Barter PJ, Rye KA. Inhibition of inflammatory signaling pathways in 3T3-L1 adipocytes by apolipoprotein A-I. FASEB J 2016; 30:2324-35. [PMID: 26965683 DOI: 10.1096/fj.201500026r] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/22/2016] [Indexed: 01/04/2023]
Abstract
Activation of inflammatory signaling pathways links obesity with metabolic disorders. TLR4-mediated activation of MAPKs and NF-κB are 2 such pathways implicated in obesity-induced inflammation. Apolipoprotein A-I (apoA-I) exerts anti-inflammatory effects on adipocytes by effluxing cholesterol from the cells via the ATP binding cassette transporter A1 (ABCA1). It is not known if these effects involve inhibition of inflammatory signaling pathways by apoA-I. This study asks if apoA-I inhibits activation of MAPKs and NF-κB in mouse 3T3-L1 adipocytes and whether this inhibition is ABCA1 dependent. Incubation of differentiated 3T3-L1 adipocytes with apoA-I decreased cell surface expression of TLR4 by 16 ± 2% and synthesis of the TLR4 adaptor protein, myeloid differentiation primary response 88, by 24 ± 4% in an ABCA1-dependent manner. ApoA-I also inhibited downstream activation of MAPKs, such as ERK, p38MAPK, and JNK, as well as expression of proinflammatory adipokines in bacterial LPS-stimulated 3T3-L1 adipocytes in an ABCA1-dependent manner. ApoA-I, by contrast, suppressed nuclear localization of the p65 subunit of NF-κB by 30 ± 3% in LPS-stimulated 3T3-L1 adipocytes in an ABCA1-independent manner. In conclusion, apoA-I inhibits TLR4-mediated inflammatory signaling pathways in adipocytes by preventing MAPK and NF-κB activation.-Sultana, A., Cochran, B. J., Tabet, F., Patel, M., Cuesta Torres, L., Barter, P. J., Rye, K.-A. Inhibition of inflammatory signaling pathways in 3T3-L1 adipocytes by apolipoprotein A-I.
Collapse
Affiliation(s)
- Afroza Sultana
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia; and
| | - Blake J Cochran
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia; and
| | - Fatiha Tabet
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia; and
| | - Mili Patel
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia; and
| | - Luisa Cuesta Torres
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia; and
| | - Philip J Barter
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia; and Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia; and Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
21
|
Nguyen SD, Maaninka K, Lappalainen J, Nurmi K, Metso J, Öörni K, Navab M, Fogelman AM, Jauhiainen M, Lee-Rueckert M, Kovanen PT. Carboxyl-Terminal Cleavage of Apolipoprotein A-I by Human Mast Cell Chymase Impairs Its Anti-Inflammatory Properties. Arterioscler Thromb Vasc Biol 2015; 36:274-84. [PMID: 26681753 PMCID: PMC4725095 DOI: 10.1161/atvbaha.115.306827] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 11/18/2015] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Apolipoprotein A-I (apoA-I) has been shown to possess several atheroprotective functions, including inhibition of inflammation. Protease-secreting activated mast cells reside in human atherosclerotic lesions. Here we investigated the effects of the neutral proteases released by activated mast cells on the anti-inflammatory properties of apoA-I. APPROACH AND RESULTS Activation of human mast cells triggered the release of granule-associated proteases chymase, tryptase, cathepsin G, carboxypeptidase A, and granzyme B. Among them, chymase cleaved apoA-I with the greatest efficiency and generated C-terminally truncated apoA-I, which failed to bind with high affinity to human coronary artery endothelial cells. In tumor necrosis factor-α-activated human coronary artery endothelial cells, the chymase-cleaved apoA-I was unable to suppress nuclear factor-κB-dependent upregulation of vascular cell adhesion molecule-1 (VCAM-1) and to block THP-1 cells from adhering to and transmigrating across the human coronary artery endothelial cells. Chymase-cleaved apoA-I also had an impaired ability to downregulate the expression of tumor necrosis factor-α, interleukin-1β, interleukin-6, and interleukin-8 in lipopolysaccharide-activated GM-CSF (granulocyte-macrophage colony-stimulating factor)- and M-CSF (macrophage colony-stimulating factor)-differentiated human macrophage foam cells and to inhibit reactive oxygen species formation in PMA (phorbol 12-myristate 13-acetate)-activated human neutrophils. Importantly, chymase-cleaved apoA-I showed reduced ability to inhibit lipopolysaccharide-induced inflammation in vivo in mice. Treatment with chymase blocked the ability of the apoA-I mimetic peptide L-4F, but not of the protease-resistant D-4F, to inhibit proinflammatory gene expression in activated human coronary artery endothelial cells and macrophage foam cells and to prevent reactive oxygen species formation in activated neutrophils. CONCLUSIONS The findings identify C-terminal cleavage of apoA-I by human mast cell chymase as a novel mechanism leading to loss of its anti-inflammatory functions. When targeting inflamed protease-rich atherosclerotic lesions with apoA-I, infusions of protease-resistant apoA-I might be the appropriate approach.
Collapse
Affiliation(s)
- Su Duy Nguyen
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Katariina Maaninka
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Jani Lappalainen
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Katariina Nurmi
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Jari Metso
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Katariina Öörni
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Mohamad Navab
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Alan M Fogelman
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Matti Jauhiainen
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Miriam Lee-Rueckert
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.)
| | - Petri T Kovanen
- From the Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland (S.D.N., K.M., J.L., K.N., K.Ö., M.L.-R., P.T.K.); National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum Helsinki, Helsinki, Finland (J.M., M.J.); and Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (M.N., A.M.F.).
| |
Collapse
|
22
|
Spillmann F, Trimpert C, Peng J, Eckerle LG, Staudt A, Warstat K, Felix SB, Pieske B, Tschöpe C, Van Linthout S. High-density lipoproteins reduce palmitate-induced cardiomyocyte apoptosis in an AMPK-dependent manner. Biochem Biophys Res Commun 2015; 466:272-7. [PMID: 26362182 DOI: 10.1016/j.bbrc.2015.09.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/05/2015] [Indexed: 12/18/2022]
Abstract
Palmitate has been implicated in the induction of cardiomyocyte apoptosis via reducing the activity of 5' AMP-activated protein kinase (AMPK). We sought to evaluate whether high-density lipoproteins (HDLs), known for their cardioprotective features and their potential to increase AMPK activity, can reduce palmitate-induced cardiomyocyte apoptosis and whether this effect is AMPK-dependent. Therefore, cardiomyocytes were isolated from adult Wistar rat hearts via perfusion on a Langendorff-apparatus and cultured in free fatty acid-free BSA control medium or 0.5 mM palmitate medium in the presence or absence of HDL (5 μg protein/ml) with or without 0.1 μM of the AMPK-inhibitor compound S for the analysis of Annexin V/propidium, genes involved in apoptosis and fatty acid oxidation, and cardiomyocyte contractility. We found that HDLs decreased palmitate-induced cardiomyocyte apoptosis as indicated by a reduction in Annexin V-positive cardiomyocytes and an increase in Bcl-2 versus Bax ratio. Concomitantly, HDLs increased the palmitate-impaired expression of genes involved in fatty acid oxidation. Furthermore, HDLs improved the palmitate-impaired cardiomyocyte contractility. All effects were mediated in an AMPK-dependent manner, concluding that HDLs reduce palmitate-induced cardiomyocyte apoptosis, resulting in improved cardiomyocyte contractility through a mechanism involving AMPK.
Collapse
Affiliation(s)
- Frank Spillmann
- Charité-University-Medicine Berlin, Campus Virchow Klinikum, Department of Cardiology, Berlin, Germany
| | - Christiane Trimpert
- Department of Internal Medicine I, University Medicine Greifswald, Greifswald, Germany
| | - Jun Peng
- Charité-University-Medicine Berlin, Campus Virchow Klinikum, Department of Cardiology, Berlin, Germany
| | - Lars G Eckerle
- Department of Internal Medicine I, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Staudt
- Department of Internal Medicine I, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Warstat
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Stephan B Felix
- Department of Internal Medicine I, University Medicine Greifswald, Greifswald, Germany; Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Greifswald, Germany
| | - Burkert Pieske
- Charité-University-Medicine Berlin, Campus Virchow Klinikum, Department of Cardiology, Berlin, Germany; Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany
| | - Carsten Tschöpe
- Charité-University-Medicine Berlin, Campus Virchow Klinikum, Department of Cardiology, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany; Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany
| | - Sophie Van Linthout
- Charité-University-Medicine Berlin, Campus Virchow Klinikum, Department of Cardiology, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany; Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany.
| |
Collapse
|
23
|
Spillmann F, Miteva K, Pieske B, Tschöpe C, Van Linthout S. High-density lipoproteins reduce endothelial-to-mesenchymal transition. Arterioscler Thromb Vasc Biol 2015; 35:1774-7. [PMID: 26088574 DOI: 10.1161/atvbaha.115.305887] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 05/26/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Endothelial-to-mesenchymal transition is an inflammation-induced process by which endothelial cells can transdifferentiate into fibroblasts. Based on the endothelial-protective and antifibrotic effects of high-density lipoproteins (HDL), we aimed to investigate whether HDL can reduce endothelial-to-mesenchymal transition. APPROACH AND RESULTS Therefore, human aortic endothelial cells were stimulated with the profibrotic factor transforming growth factor (TGF)-β1 in the presence or absence of HDL. Their impact on the transition of endothelial cells to mesenchymal-like cells was analyzed. Phase contrast microscopy demonstrated that HDL abrogated the TGF-β1-induced spindle-shape morphology in human aortic endothelial cells. Furthermore, HDL decreased the TGF-β1-mediated induction of α-smooth muscle actin expression and concomitant loss in endothelial cadherin expression, as shown by immunofluorescence staining and flow cytometry. In addition, HDL decreased the TGF-β1-induced collagen deposition in human aortic endothelial cells involving the scavenger receptor class B, type 1 and downstream phosphatidyl inositol-3-kinase following the findings that the HDL-mediated reduction was abrogated by scavenger receptor class B, type 1 siRNA knockdown and phosphatidyl inositol-3-kinase inhibition, respectively. The HDL-mediated reduction in endothelial-to-mesenchymal transition was associated with an induction of the inhibitory Smad, Smad 7. CONCLUSIONS We provide the first in vitro evidence that the endothelial-protective and antifibrotic effects of HDL include the reduction in endothelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Frank Spillmann
- From the Department of Cardiology (F.S., B.P., C.T.) and Berlin-Brandenburg Center for Regenerative Therapy (BCRT) (K.M., C.T., S.V.L.), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany; Department of Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany (B.P.); and Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany (B.P., C.T., S.V.L.)
| | - Kapka Miteva
- From the Department of Cardiology (F.S., B.P., C.T.) and Berlin-Brandenburg Center for Regenerative Therapy (BCRT) (K.M., C.T., S.V.L.), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany; Department of Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany (B.P.); and Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany (B.P., C.T., S.V.L.)
| | - Burkert Pieske
- From the Department of Cardiology (F.S., B.P., C.T.) and Berlin-Brandenburg Center for Regenerative Therapy (BCRT) (K.M., C.T., S.V.L.), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany; Department of Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany (B.P.); and Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany (B.P., C.T., S.V.L.)
| | - Carsten Tschöpe
- From the Department of Cardiology (F.S., B.P., C.T.) and Berlin-Brandenburg Center for Regenerative Therapy (BCRT) (K.M., C.T., S.V.L.), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany; Department of Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany (B.P.); and Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany (B.P., C.T., S.V.L.)
| | - Sophie Van Linthout
- From the Department of Cardiology (F.S., B.P., C.T.) and Berlin-Brandenburg Center for Regenerative Therapy (BCRT) (K.M., C.T., S.V.L.), Charité-University-Medicine Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany; Department of Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany (B.P.); and Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Germany (B.P., C.T., S.V.L.).
| |
Collapse
|
24
|
Van Linthout S, Frias M, Singh N, De Geest B. Therapeutic potential of HDL in cardioprotection and tissue repair. Handb Exp Pharmacol 2015; 224:527-565. [PMID: 25523001 DOI: 10.1007/978-3-319-09665-0_17] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Epidemiological studies support a strong association between high-density lipoprotein (HDL) cholesterol levels and heart failure incidence. Experimental evidence from different angles supports the view that low HDL is unlikely an innocent bystander in the development of heart failure. HDL exerts direct cardioprotective effects, which are mediated via its interactions with the myocardium and more specifically with cardiomyocytes. HDL may improve cardiac function in several ways. Firstly, HDL may protect the heart against ischaemia/reperfusion injury resulting in a reduction of infarct size and thus in myocardial salvage. Secondly, HDL can improve cardiac function in the absence of ischaemic heart disease as illustrated by beneficial effects conferred by these lipoproteins in diabetic cardiomyopathy. Thirdly, HDL may improve cardiac function by reducing infarct expansion and by attenuating ventricular remodelling post-myocardial infarction. These different mechanisms are substantiated by in vitro, ex vivo, and in vivo intervention studies that applied treatment with native HDL, treatment with reconstituted HDL, or human apo A-I gene transfer. The effect of human apo A-I gene transfer on infarct expansion and ventricular remodelling post-myocardial infarction illustrates the beneficial effects of HDL on tissue repair. The role of HDL in tissue repair is further underpinned by the potent effects of these lipoproteins on endothelial progenitor cell number, function, and incorporation, which may in particular be relevant under conditions of high endothelial cell turnover. Furthermore, topical HDL therapy enhances cutaneous wound healing in different models. In conclusion, the development of HDL-targeted interventions in these strategically chosen therapeutic areas is supported by a strong clinical rationale and significant preclinical data.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Charité-University-Medicine Berlin, Campus Virchow, Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Berlin, Germany
| | | | | | | |
Collapse
|
25
|
The Impact of Lipoproteins on Wound Healing: Topical HDL Therapy Corrects Delayed Wound Healing in Apolipoprotein E Deficient Mice. Pharmaceuticals (Basel) 2014; 7:419-32. [PMID: 24705596 PMCID: PMC4014700 DOI: 10.3390/ph7040419] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/06/2014] [Accepted: 03/26/2014] [Indexed: 12/22/2022] Open
Abstract
Chronic non-healing wounds lead to considerable morbidity and mortality. Pleiotropic effects of high density lipoproteins (HDL) may beneficially affect wound healing. The objectives of this murine study were: (1) to investigate the hypothesis that hypercholesterolemia induces impaired wound healing and (2) to study the effect of topical HDL administration in a model of delayed wound healing. A circular full thickness wound was created on the back of each mouse. A silicone splint was used to counteract wound contraction. Coverage of the wound by granulation tissue and by epithelium was quantified every 2 days. Re-epithelialization from day 0 till day 10 was unexpectedly increased by 21.3% (p < 0.05) in C57BL/6 low density lipoprotein (LDLr) deficient mice with severe hypercholesterolemia (489 ± 14 mg/dL) compared to C57BL/6 mice and this effect was entirely abrogated following cholesterol lowering adenoviral LDLr gene transfer. In contrast, re-epithelialization in hypercholesterolemic (434 ± 16 mg/dL) C57BL/6 apolipoprotein (apo) E−/− mice was 22.6% (p < 0.0001) lower than in C57BL/6 mice. Topical HDL gel administered every 2 days increased re-epithelialization by 25.7% (p < 0.01) in apo E−/− mice. In conclusion, topical HDL application is an innovative therapeutic strategy that corrects impaired wound healing in apo E−/− mice.
Collapse
|
26
|
LXR agonism improves TNF-α-induced endothelial dysfunction in the absence of its cholesterol-modulating effects. Atherosclerosis 2014; 232:1-9. [DOI: 10.1016/j.atherosclerosis.2013.10.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 09/13/2013] [Accepted: 10/01/2013] [Indexed: 12/20/2022]
|
27
|
Wu BJ, Ong KL, Shrestha S, Chen K, Tabet F, Barter PJ, Rye KA. Inhibition of arthritis in the Lewis rat by apolipoprotein A-I and reconstituted high-density lipoproteins. Arterioscler Thromb Vasc Biol 2013; 34:543-51. [PMID: 24357062 DOI: 10.1161/atvbaha.113.302832] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE This study questions whether high-density lipoproteins (HDLs) and apolipoprotein A-I inhibit joint inflammation in streptococcal cell wall peptidoglycan-polysaccharide (PG-PS)-induced arthritis in female Lewis rats. APPROACH AND RESULTS Administration of PG-PS to female Lewis rats caused acute joint inflammation after 4 days, followed by remission by day 8. The animals subsequently developed chronic joint inflammation that persisted until euthanasia at day 21. Treatment with apolipoprotein A-I 24 hours before and 24 hours after PG-PS administration reduced the acute and chronic joint inflammation. Treatment with apolipoprotein A-I at days 7, 9, and 11 after PG-PS administration reduced the chronic joint inflammation. Treatment with apolipoprotein A-I or reconstituted HDLs consisting of apolipoprotein A-I complexed with phosphatidylcholine 24 hours before and at days 1, 7, 9, and 11 after PG-PS administration reduced acute and chronic joint inflammation. Treatment with apolipoprotein A-I also reduced the inflammatory white blood cell count, synovial fluid proinflammatory cytokine levels, synovial tissue macrophage accumulation, as well as toll-like receptor 2, and inflammatory cytokine expression. At the molecular level, preincubation of human monocyte-derived macrophages with apolipoprotein A-I or reconstituted HDLs before PG-PS stimulation inhibited the PG-PS-induced increase in toll-like receptor 2 and myeloid differentiation primary response gene (88) mRNA levels, nuclear factor-κB activation, and proinflammatory cytokine production. The effects of apolipoprotein A-I and reconstituted HDLs were abolished by transfecting the human monocyte-derived macrophages with ATP-binding cassette transporter A1 or G1 siRNA. CONCLUSIONS Apolipoprotein A-I and reconstituted HDLs attenuate PG-PS-induced arthritis in the rat. Studies in human monocyte-derived macrophages indicate that this benefit may be because of the inhibition of toll-like receptor 2 expression and decreased nuclear factor-κB activation in macrophages.
Collapse
Affiliation(s)
- Ben J Wu
- From the Lipid Research Group, The Heart Research Institute, Sydney, New South Wales, Australia (B.J.W., K.L.O., S.S., K.C., F.T., P.J.B., K.-A.R.); Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia (B.J.W., K.L.O., F.T., P.J.B., K.-A.R.); and Centre for Vascular Research, The University of New South Wales, Sydney, New South Wales, Australia (B.J.W., K.L.O., S.S., F.T., P.J.B., K.-A.R.)
| | | | | | | | | | | | | |
Collapse
|
28
|
Stancu CS, Toma L, Sima AV. Dual role of lipoproteins in endothelial cell dysfunction in atherosclerosis. Cell Tissue Res 2012; 349:433-46. [DOI: 10.1007/s00441-012-1437-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 04/12/2012] [Indexed: 12/28/2022]
|
29
|
Cheng AM, Handa P, Tateya S, Schwartz J, Tang C, Mitra P, Oram JF, Chait A, Kim F. Apolipoprotein A-I attenuates palmitate-mediated NF-κB activation by reducing Toll-like receptor-4 recruitment into lipid rafts. PLoS One 2012; 7:e33917. [PMID: 22479476 PMCID: PMC3316516 DOI: 10.1371/journal.pone.0033917] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 02/20/2012] [Indexed: 11/24/2022] Open
Abstract
While high-density lipoprotein (HDL) is known to protect against a wide range of inflammatory stimuli, its anti-inflammatory mechanisms are not well understood. Furthermore, HDL's protective effects against saturated dietary fats have not been previously described. In this study, we used endothelial cells to demonstrate that while palmitic acid activates NF-κB signaling, apolipoprotein A–I, (apoA-I), the major protein component of HDL, attenuates palmitate-induced NF-κB activation. Further, vascular NF-κB signaling (IL-6, MCP-1, TNF-α) and macrophage markers (CD68, CD11c) induced by 24 weeks of a diabetogenic diet containing cholesterol (DDC) is reduced in human apoA-I overexpressing transgenic C57BL/6 mice compared to age-matched WT controls. Moreover, WT mice on DDC compared to a chow diet display increased gene expression of lipid raft markers such as Caveolin-1 and Flotillin-1, and inflammatory Toll-like receptors (TLRs) (TLR2, TLR4) in the vasculature. However apoA-I transgenic mice on DDC show markedly reduced expression of these genes. Finally, we show that in endothelial cells TLR4 is recruited into lipid rafts in response to palmitate, and that apoA-I prevents palmitate-induced TLR4 trafficking into lipid rafts, thereby blocking NF-κB activation. Thus, apoA-I overexpression might be a useful therapeutic tool against vascular inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Francis Kim
- Department of Medicine, Diabetes and Obesity Center of Excellence, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
30
|
Regression and stabilization of advanced murine atherosclerotic lesions: a comparison of LDL lowering and HDL raising gene transfer strategies. J Mol Med (Berl) 2011; 89:555-67. [PMID: 21249329 PMCID: PMC3098380 DOI: 10.1007/s00109-011-0722-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Revised: 12/04/2010] [Accepted: 12/30/2010] [Indexed: 11/05/2022]
Abstract
Both reductions in atherogenic lipoproteins and increases in high-density lipoprotein (HDL) levels may affect atherosclerosis regression. Here, the relative potential of low-density lipoprotein (LDL) lowering and HDL raising gene transfer strategies to induce regression of complex murine atherosclerotic lesions was directly compared. Male C57BL/6 LDL receptor (LDLr)−/− mice were fed an atherogenic diet (1.25% cholesterol and 10% coconut oil) to induce advanced atherosclerotic lesions. A baseline group was killed after 6 months and remaining mice were randomized into a control progression (Adnull or saline), an apolipoprotein (apo) A-I (AdA-I), an LDLr (AdLDLr), or a combined apo A-I/LDLr (AdA-I/AdLDLr) adenoviral gene transfer group and followed-up for another 12 weeks with continuation of the atherogenic diet. Gene transfer with AdLDLr decreased non-HDL cholesterol levels persistently by 95% (p < 0.001) compared with baseline. This drastic reduction of non-HDL cholesterol levels induced lesion regression by 28% (p < 0.001) in the aortic root and by 25% (p < 0.05) in the brachiocephalic artery at 12 weeks after transfer. Change in lesion size was accompanied by enhanced plaque stability, as evidenced by increased collagen content, reduced lesional macrophage content, a drastic reduction of necrotic core area, and decreased expression of inflammatory genes. Elevated HDL cholesterol following AdA-I transfer increased collagen content in lesions, but did not induce regression. Apo A-I gene transfer on top of AdLDLr transfer resulted in additive effects, particularly on inflammatory gene expression. In conclusion, drastic lipid lowering induced by a powerful gene transfer strategy leads to pronounced regression and stabilization of advanced murine atherosclerosis.
Collapse
|
31
|
Chen X, Wu X, Zhao Y, Wang G, Feng J, Li Q, Qian G. A novel binding protein of single immunoglobulin IL-1 receptor-related molecule: Paralemmin-3. Biochem Biophys Res Commun 2010; 404:1029-33. [PMID: 21187075 DOI: 10.1016/j.bbrc.2010.12.104] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 12/21/2010] [Indexed: 10/18/2022]
Abstract
Previous studies have shown that single immunoglobulin IL-1 receptor-related molecule (SIGIRR) is a negative regulator of Toll-Interleukin-1 receptor signaling. Nevertheless, the molecular mechanism of the negatively regulatory effect of SIGIRR remains unknown. Using a yeast two-hybrid screen, we identified paralemmin-3 (PALM3) as a novel binding protein of SIGIRR. This interaction of SIGIRR with PALM3 was confirmed by coimmunoprecipitation in mammalian cells. In addition, the PALM3 mRNA expression was upregulated by lipopolysaccharide (LPS)-stimulation in a human alveolar epithelial cell line (A549 cells). Furthermore, silencing PALM3 by RNA interference inhibited the release of inflammatory cytokines in A549 cells after LPS-stimulation. These results suggest that PALM3 may function as an adaptor in the LPS- Toll-like receptor 4 signaling and the interaction of SIGIRR with PALM3 may partly account for the mechanism of the negatively regulatory effect of SIGIRR.
Collapse
Affiliation(s)
- Xuxin Chen
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, PR China
| | | | | | | | | | | | | |
Collapse
|