Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Publication year range
1.
Am J Physiol Cell Physiol ; 324(2): C438-C446, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36534503

ABSTRACT

Apolipoprotein A-I (apoA-I) mediates reverse cholesterol transport (RCT) out of cells. In addition to its important role in the RTC, apoA-I also possesses anti-inflammatory and antioxidative functions including the ability to activate inflammasome and signal via toll-like receptors. Dysfunctional apoA-I or its low abundance may cause accumulation of cholesterol mass in alveolar macrophages, leading to the formation of foam cells. Increased numbers of foam cells have been noted in the lungs of mice after experimental exposure to cigarette smoke, silica, or bleomycin and in the lungs of patients suffering from different types of lung fibrosis, including idiopathic pulmonary fibrosis (IPF). This suggests that dysregulation of lipid metabolism may be a common event in the pathogenesis of interstitial lung diseases. Recognition of the emerging role of cholesterol in the regulation of lung inflammation and remodeling provides a challenging concept for understanding lung diseases and offers novel and exciting avenues for therapeutic development. Accordingly, a number of preclinical studies demonstrated decreased expression of inflammatory and profibrotic mediators and preserved lung tissue structure following the administration of the apoA-I or its mimetic peptides. This review highlights the role of apoA-I in lung fibrosis and provides evidence for its potential use in the treatment of this pathological condition.


Subject(s)
Apolipoprotein A-I , Idiopathic Pulmonary Fibrosis , Animals , Mice , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/therapeutic use , Atherosclerosis/metabolism , Cholesterol/metabolism , Foam Cells/metabolism , Foam Cells/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism
2.
BMC Med ; 21(1): 392, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37915050

ABSTRACT

BACKGROUND: Sepsis is characterized by a dysregulated immune response and metabolic alterations, including decreased high-density lipoprotein cholesterol (HDL-C) levels. HDL exhibits beneficial properties, such as lipopolysaccharides (LPS) scavenging, exerting anti-inflammatory effects and providing endothelial protection. We investigated the effects of CER-001, an engineered HDL-mimetic, in a swine model of LPS-induced acute kidney injury (AKI) and a Phase 2a clinical trial, aiming to better understand its molecular basis in systemic inflammation and renal function. METHODS: We carried out a translational approach to study the effects of HDL administration on sepsis. Sterile systemic inflammation was induced in pigs by LPS infusion. Animals were randomized into LPS (n = 6), CER20 (single dose of CER-001 20 mg/kg; n = 6), and CER20 × 2 (two doses of CER-001 20 mg/kg; n = 6) groups. Survival rate, endothelial dysfunction biomarkers, pro-inflammatory mediators, LPS, and apolipoprotein A-I (ApoA-I) levels were assessed. Renal and liver histology and biochemistry were analyzed. Subsequently, we performed an open-label, randomized, dose-ranging (Phase 2a) study included 20 patients with sepsis due to intra-abdominal infection or urosepsis, randomized into Group A (conventional treatment, n = 5), Group B (CER-001 5 mg/kg BID, n = 5), Group C (CER-001 10 mg/kg BID, n = 5), and Group D (CER-001 20 mg/kg BID, n = 5). Primary outcomes were safety and efficacy in preventing AKI onset and severity; secondary outcomes include changes in inflammatory and endothelial dysfunction markers. RESULTS: CER-001 increased median survival, reduced inflammatory mediators, complement activation, and endothelial dysfunction in endotoxemic pigs. It enhanced LPS elimination through the bile and preserved liver and renal parenchyma. In the clinical study, CER-001 was well-tolerated with no serious adverse events related to study treatment. Rapid ApoA-I normalization was associated with enhanced LPS removal and immunomodulation with improvement of clinical outcomes, independently of the type and gravity of the sepsis. CER-001-treated patients had reduced risk for the onset and progression to severe AKI (stage 2 or 3) and, in a subset of critically ill patients, a reduced need for organ support and shorter ICU length of stay. CONCLUSIONS: CER-001 shows promise as a therapeutic strategy for sepsis management, improving outcomes and mitigating inflammation and organ damage. TRIAL REGISTRATION: The study was approved by the Agenzia Italiana del Farmaco (AIFA) and by the Local Ethic Committee (N° EUDRACT 2020-004202-60, Protocol CER-001- SEP_AKI_01) and was added to the EU Clinical Trials Register on January 13, 2021.


Subject(s)
Acute Kidney Injury , Sepsis , Humans , Animals , Swine , Lipoproteins, HDL , Apolipoprotein A-I/therapeutic use , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/pharmacology , Lipopolysaccharides , Translational Research, Biomedical , Inflammation , Sepsis/drug therapy , Acute Kidney Injury/drug therapy , Inflammation Mediators
3.
Liver Int ; 43(1): 234-248, 2023 01.
Article in English | MEDLINE | ID: mdl-36203339

ABSTRACT

BACKGROUND AND AIMS: Apolipoprotein A-1 (ApoA-1), the major apolipoprotein of high-density lipoprotein, plays anti-atherogenic role in cardiovascular diseases and exerts anti-inflammation effect in various inflammatory and infectious diseases. However, the role and mechanism of ApoA-1 in hepatic ischaemia-reperfusion (I/R) injury is unknown. METHODS: In this study, we measured ApoA-1 expression in human liver grafts after transplantation. Mice partial hepatic I/R injury model was made in ApoA-1 knockout mice, ApoA-1 mimetic peptide D-4F treatment mice and corresponding control mice to examine the effect of ApoA-1 on liver damage, inflammation response and cell death. Primary hepatocytes and macrophages were isolated for in vitro study. RESULTS: The results showed that ApoA-1 expression was down-regulated in human liver grafts after transplantation and mice livers subjected to hepatic I/R injury. ApoA-1 deficiency aggravated liver damage and inflammation response induced by hepatic I/R injury. Interestingly, we found that ApoA-1 deficiency increased pyroptosis instead of apoptosis during acute phase of hepatic I/R injury, which mainly occurred in macrophages rather than hepatocytes. The inhibition of pyroptosis compensated for the adverse impact of ApoA-1 deficiency. Furthermore, the up-regulated pyroptosis process was testified to be mediated by ApoA-1 through TLR4-NF-κB pathway and TLR4 inhibition significantly improved hepatic I/R injury. In addition, we confirmed that D-4F ameliorated hepatic I/R injury. CONCLUSIONS: Our study has identified the protective role of ApoA-1 in hepatic I/R injury through inhibiting pyroptosis in macrophages via TLR4-NF-κB pathway. The effect of ApoA-1 may provide a novel therapeutic approach for hepatic I/R injury.


Subject(s)
Liver Diseases , Reperfusion Injury , Humans , Mice , Animals , NF-kappa B/metabolism , Apolipoprotein A-I/pharmacology , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/therapeutic use , Pyroptosis , Toll-Like Receptor 4 , Signal Transduction , Liver/metabolism , Liver Diseases/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Macrophages/metabolism
4.
J Intern Med ; 291(3): 364-370, 2022 03.
Article in English | MEDLINE | ID: mdl-34761839

ABSTRACT

BACKGROUND: Kidney failure is the major cause of morbidity and mortality in familial lecithin:cholesterol acyltransferase deficiency (FLD), a rare inherited lipid disorder with no cure. Lipoprotein X (LpX), an abnormal lipoprotein, is primarily accountable for nephrotoxicity. METHODS: CER-001 was tested in an FLD patient with dramatic kidney disease for 12 weeks. RESULTS: Infusions of CER-001 normalized the lipoprotein profile, with a disappearance of the abnormal LpX in favour of normal-sized LDL. The worsening of kidney function was slowed by the treatment, and kidney biopsy showed a slight reduction of lipid deposits and a stabilization of the disease. In vitro experiments demonstrate that CER-001 progressively reverts lipid accumulation in podocytes by a dual effect: remodelling plasma lipoproteins and removing LpX-induced lipid deposit. CONCLUSION: This study demonstrates that CER-001 may represent a therapeutic option in FLD patients. It also has the potential to be beneficial in other renal diseases characterized by kidney lipid deposits.


Subject(s)
Lecithin Cholesterol Acyltransferase Deficiency , Apolipoprotein A-I/therapeutic use , Humans , Kidney/pathology , Lecithin Cholesterol Acyltransferase Deficiency/drug therapy , Lecithin Cholesterol Acyltransferase Deficiency/pathology , Lipoproteins , Phosphatidylcholine-Sterol O-Acyltransferase/pharmacology , Phosphatidylcholine-Sterol O-Acyltransferase/therapeutic use , Phospholipids , Recombinant Proteins
5.
Curr Atheroscler Rep ; 24(7): 585-597, 2022 07.
Article in English | MEDLINE | ID: mdl-35524914

ABSTRACT

PURPOSE OF REVIEW: The elevated adverse cardiovascular event rate among patients with low high-density lipoprotein cholesterol (HDL-C) formed the basis for the hypothesis that elevating HDL-C would reduce those events. Attempts to raise endogenous HDL-C levels, however, have consistently failed to show improvements in cardiovascular outcomes. However, steady-state HDL-C concentration does not reflect the function of this complex family of particles. Indeed, HDL functions correlate only weakly with serum HDL-C concentration. Thus, the field has pivoted from simply raising the quantity of HDL-C to a focus on improving the putative anti-atherosclerotic functions of HDL particles. Such functions include the ability of HDL to promote the efflux of cholesterol from cholesterol-laden macrophages. Apolipoprotein A-I (apoA-I), the signature apoprotein of HDL, may facilitate the removal of cholesterol from atherosclerotic plaque, reduce the lesional lipid content and might thus stabilize vulnerable plaques, thereby reducing the risk of cardiac events. Infusion of preparations of apoA-I may improve cholesterol efflux capacity (CEC). This review summarizes the development of apoA-I therapies, compares their structural and functional properties and discusses the findings of previous studies including their limitations, and how CSL112, currently being tested in a phase III trial, may overcome these challenges. RECENT FINDINGS: Three major ApoA-I-based approaches (MDCO-216, CER-001, and CSL111/CSL112) have aimed to enhance reverse cholesterol transport. These three therapies differ considerably in both lipid and protein composition. MDCO-216 contains recombinant ApoA-I Milano, CER-001 contains recombinant wild-type human ApoA-I, and CSL111/CSL112 contains native ApoA-I isolated from human plasma. Two of the three agents studied to date (apoA-1 Milano and CER-001) have undergone evaluation by intravascular ultrasound imaging, a technique that gauges lesion volume well but does not assess other important variables that may relate to clinical outcomes. ApoA-1 Milano and CER-001 reduce lecithin-cholesterol acyltransferase (LCAT) activity, potentially impairing the function of HDL in reverse cholesterol transport. Furthermore, apoA-I Milano can compete with and alter the function of the recipient's endogenous apoA-I. In contrast to these agents, CSL112, a particle formulated using human plasma apoA-I and phosphatidylcholine, increases LCAT activity and does not lead to the malfunction of endogenous apoA-I. CSL112 robustly increases cholesterol efflux, promotes reverse cholesterol transport, and now is being tested in a phase III clinical trial. Phase II-b studies of MDCO-216 and CER-001 failed to produce a significant reduction in coronary plaque volume as assessed by IVUS. However, the investigation to determine whether the direct infusion of a reconstituted apoA-I reduces post-myocardial infarction coronary events is being tested using CSL112, which is dosed at a higher level than MDCO-216 and CER-001 and has more favorable pharmacodynamics.


Subject(s)
Acute Coronary Syndrome , Atherosclerosis , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/therapeutic use , Atherosclerosis/drug therapy , Cholesterol/metabolism , Cholesterol, HDL , Humans
6.
Heart Vessels ; 37(12): 2128-2136, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35739432

ABSTRACT

We aimed to evaluate the impact of biologic treatment on subclinical atherosclerosis and risk factors for cardiovascular disease (CVD) in patients with rheumatoid arthritis (RA). Forty-nine biologic naïve RA patients, treated with conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs), who were eligible for treatment with a biologic agent, were included in the study. The serum levels of lipid parameters, as well as disease activity parameters were determined in RA patients before and after 3 and 6 months of therapy. Carotid artery intima-media thickness (cIMT) was measured before and after treatment. A comparison analysis of change of these parameters was also performed between anti-tumor necrosis factor (anti-TNF) and non-anti-TNF users. Furthermore, 31 non-smoking healthy volunteers, matched for age and gender, were used as a control group. At baseline, RA patients had a decrease in serum total cholesterol (TC) and high-density lipoprotein cholesterol (HDL-C) levels compared with controls (209 ± 63 vs 233 ± 44 and 58 ± 15 vs 61 ± 14, p < 0.004), while cIMT was higher versus controls [0.9 (0.8-1) vs 0.6 (0.5-0.7), p < 0.001]. TC, HDL-C and apolipoprotein A1 levels were significantly increased 3 months after treatment (209 ± 63, 58 ± 15, 162 ± 32, vs 227 ± 45, 60 ± 15, 169 ± 29, respectively, p < 0.03) and this observation remained stable at a 6-month follow-up. After 6 months, there was also a statistically significant decrease in the cIMT [0.9 (0.8-1) vs 0.7 (0.6-0.8), p < 0.001]. Anti-TNF and non-anti-TNF users had comparable changes in cardiovascular risk parameters. The atherogenic lipid profile and subclinical atherosclerosis are features of RA, which appeared improved after biologic therapy initiation.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Atherosclerosis , Biological Products , Cardiovascular Diseases , Humans , Antirheumatic Agents/therapeutic use , Apolipoprotein A-I/therapeutic use , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Atherosclerosis/diagnosis , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Biological Products/pharmacology , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Carotid Intima-Media Thickness , Cholesterol , Heart Disease Risk Factors , Lipoproteins, HDL , Risk Factors , Tumor Necrosis Factor-alpha
7.
Adv Exp Med Biol ; 1377: 171-187, 2022.
Article in English | MEDLINE | ID: mdl-35575930

ABSTRACT

A wealth of evidence indicates that high-density lipoprotein assumes the unique antiatherosclerosis and other cardioprotective properties. Based on that, HDL-C has been considered as a promising therapy target to reduce the cardiovascular diseases. Recombinant HDL (rHDL) and apolipoprotein mimetic peptides emerge in recent years and have great potential in the future. Here we discussed the pleiotropic therapeutic effect of rHDL based on the effects of atherogenic, angiogenesis, platelet, vascular, and Alzheimer's disease. On the other hand, rHDL not only plays the key role as the major protein component of HDL, it is also used as a nanovector in antiatherosclerotic, antitumor, cardiovascular diagnosing and other therapeutic areas. Synthetic apolipoprotein mimetic peptides like apoA-I and and apoE mimetics have undergone clinical assessment, and we have also reviewed the advances of clinical trials and gave an outlook for the therapy of rHDL and mimetic peptides.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/therapeutic use , Apolipoproteins , Atherosclerosis/drug therapy , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Humans , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/therapeutic use , Peptides/therapeutic use
8.
J Immunol ; 203(3): 696-704, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31209101

ABSTRACT

Multiple sclerosis (MS) is a chronic autoimmune disease with no curative treatment. The immune regulatory properties of type I IFNs have led to the approval of IFN-ß for the treatment of relapsing-remitting MS. However, there is still an unmet need to improve the tolerability and efficacy of this therapy. In this work, we evaluated the sustained delivery of IFN-α1, either alone or fused to apolipoprotein A-1 by means of an adeno-associated viral (AAV) system in the mouse model of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis. These in vivo experiments demonstrated the prophylactic and therapeutic efficacy of the AAV-IFN-α or AAV-IFN-α fused to apolipoprotein A-1 vectors in experimental autoimmune encephalomyelitis, even at low doses devoid of hematological or neurologic toxicity. The sustained delivery of such low-dose IFN-α resulted in immunomodulatory effects, consisting of proinflammatory monocyte and T regulatory cell expansion. Moreover, encephalitogenic T lymphocytes from IFN-α-treated mice re-exposed to the myelin oligodendrocyte glycoprotein peptide in vitro showed a reduced proliferative response and cytokine (IL-17A and IFN-γ) production, in addition to upregulation of immunosuppressive molecules, such as IL-10, IDO, or PD-1. In conclusion, the results of the present work support the potential of sustained delivery of low-dose IFN-α for the treatment of MS and likely other T cell-dependent chronic autoimmune disorders.


Subject(s)
Apolipoprotein A-I/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon-alpha/therapeutic use , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-17/biosynthesis , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Programmed Cell Death 1 Receptor/biosynthesis , T-Lymphocytes, Regulatory/cytology
9.
J Pharmacol Exp Ther ; 372(1): 54-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31649050

ABSTRACT

Obesity is a pathologic condition generated by an energy imbalance, that is, excess caloric consumption, leading to weight gain and metabolic disturbances characterized by adipose tissue inflammation and hyperglycemic conditions. In line with these observations, increasing evidence causally links inflammation, or the molecules and networks integral to inflammatory response, to the development of obesity and the complications that emerge from this pathology, such as cardiovascular, neurologic, respiratory, and metabolic illnesses, as well as sepsis and cancer. Not surprisingly, this chronic and abnormal metabolic background leads to constant derangements in innate and adaptive immunity. It is well known that high-density lipoprotein (HDL) possesses anti-inflammatory and antioxidant properties, and various studies have highlighted an emerging role of HDL in modulating immune function. The efficacy of synthetic HDL (sHDL) containing the recombinant form of apoA-IMilano (sHDL-apoA-IM), originating from the observation that carriers of this mutation have low levels of HDL cholesterol without increased atherosclerosis, has been largely proved in diverse animal models of atherosclerosis; however, the therapeutic use of sHDL-apoA-IM still needs clinical validation. One of the main limitations to the use of recombinant proteins in clinical studies lies in the unsustainable purification costs. Unpurified rice-milk-apoA-IM demonstrated anti-inflammatory and antiatherogenic properties in a mouse model, even though administrated by an unconventional way: by oral gavage. Additionally, recent data have uncovered new therapeutic applications for this sHDL-apoA-IM This review provides an overview of all potential application of sHDL-apoA-IM in some inflammatory-based diseases. SIGNIFICANCE STATEMENT: A recent study demonstrated that oral administration of rice-seed protein extracts containing the apoA-IM (i.e., the milk-apoA-IM) reduced atherosclerosis development in a mouse model. Moreover, the rice-milk-apoA-IM preserved both in vitro and in vivo anti-inflammatory properties, as observed when sHDL-apoA-IM was given by intravascular infusion. Besides, various studies suggested that sHDL-apoA-IM could positively affect other inflammatory-based diseases. Together, these data might represent a new starting point for "sHDL-apoA-IM-based therapies" in chronic degenerative disease.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Anticholesteremic Agents/therapeutic use , Apolipoprotein A-I/therapeutic use , Atherosclerosis/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Anticholesteremic Agents/administration & dosage , Apolipoprotein A-I/administration & dosage , Atherosclerosis/prevention & control , Humans
10.
Arterioscler Thromb Vasc Biol ; 38(4): 953-963, 2018 04.
Article in English | MEDLINE | ID: mdl-29437574

ABSTRACT

OBJECTIVE: CSL112 (apolipoprotein A-I [apoA-I; human]) is a novel formulation of apoA-I in development for reduction of early recurrent cardiovascular events after acute myocardial infarction. Cholesterol efflux capacity (CEC) is a marker of high-density lipoprotein (HDL) function that is strongly correlated with incident cardiovascular disease. Impaired CEC has been observed in patients with coronary heart disease. Here, we determined whether infused apoA-I improves CEC when administered to patients with stable atherosclerotic disease versus healthy volunteers. APPROACH AND RESULTS: Measurements of apoA-I, HDL unesterified cholesterol, HDL esterified cholesterol, pre-ß1-HDL, and CEC were determined in samples from patients with stable atherosclerotic disease before and after intravenous administration of CSL112. These measures were compared with 2 prior studies in healthy volunteers for differences in CEC at baseline and after CSL112 infusion. Patients with stable atherosclerotic disease exhibited significantly lower ATP-binding cassette transporter 1-mediated CEC at baseline (P<0.0001) despite slightly higher apoA-I levels when compared with healthy individuals (2 phase 1 studies pooled; P≤0.05), suggesting impaired HDL function. However, no differences were observed in apoA-I pharmacokinetics or in pre-ß1-HDL (P=0.5) or CEC (P=0.1) after infusion of CSL112. Similar elevation in CEC was observed in patients with low or high baseline HDL function (based on tertiles of apoA-I-normalized CEC; P=0.1242). These observations were extended and confirmed using cholesterol esterification as an additional measure. CONCLUSIONS: CSL112 shows comparable, strong, and immediate effects on CEC despite underlying cardiovascular disease. CSL112 is, therefore, a promising novel therapy for lowering the burden of atherosclerosis and reducing the risk of recurrent cardiovascular events.


Subject(s)
Anticholesteremic Agents/therapeutic use , Apolipoprotein A-I/therapeutic use , Atherosclerosis/drug therapy , Cholesterol/blood , Lipid Metabolism/drug effects , Lipoproteins, HDL/therapeutic use , Adolescent , Adult , Aged , Anticholesteremic Agents/blood , Anticholesteremic Agents/pharmacokinetics , Apolipoprotein A-I/blood , Apolipoprotein A-I/pharmacokinetics , Atherosclerosis/blood , Atherosclerosis/diagnosis , Biomarkers/blood , Cholesterol, HDL/blood , Female , Healthy Volunteers , High-Density Lipoproteins, Pre-beta/blood , Humans , Lipoproteins, HDL/blood , Lipoproteins, HDL/pharmacokinetics , Male , Middle Aged , Queensland , South Australia , Treatment Outcome , United States , Young Adult
11.
Arterioscler Thromb Vasc Biol ; 38(8): 1691-1701, 2018 08.
Article in English | MEDLINE | ID: mdl-29954755

ABSTRACT

Revascularization because of coronary artery disease is commonly achieved by percutaneous coronary intervention with stent deployment. Refinement in interventional techniques, major improvements in stent design (particularly drug-eluting stents), and adjunctive pharmacotherapy with dual antiplatelet regimens have led to marked reductions in the overall rates of stent failure. However, even with the advancements made in the latest generation of drug-eluting stents, unresolved biological problems persist including delayed re-endothelialization and neoatherosclerosis, which can promote late expansion of the neointima and late stent thrombosis. Novel strategies are still needed beyond what is currently available to specifically address the pathobiological processes that underpin the residual risk for adverse clinical events. This review focuses on the emerging evidence that HDL (high-density lipoproteins) and its main apo (apolipoprotein), apoA-I, exhibit multiple vascular biological functions that are associated with an improvement in stent biocompatibility. HDL/apoA-I have recently been shown to inhibit in-stent restenosis in animal models of stenting and suppress smooth muscle cell proliferation in in vitro studies. Reconstituted HDL also promotes endothelial cell migration, endothelial progenitor cell mobilization, and re-endothelialization. Furthermore, reconstituted HDL decreases platelet activation and HDL cholesterol is inversely associated with the risk of thrombosis. Finally, reconstituted HDL/apoA-I suppresses key inflammatory mechanisms that initiate in-stent neoatherosclerosis and can efflux cholesterol from plaque macrophages, an important function of HDLs that prevents plaque progression. These unique multifunctional effects of HDL/apoA-I suggest that, if translated appropriately, have the potential to improve stent biocompatibility. This may provide an alternate and more efficacious therapeutic pathway for the translation of HDL.


Subject(s)
Apolipoprotein A-I/therapeutic use , Coronary Artery Disease/surgery , Coronary Vessels/drug effects , Coronary Vessels/surgery , Lipoproteins, HDL/therapeutic use , Percutaneous Coronary Intervention/instrumentation , Re-Epithelialization/drug effects , Stents , Animals , Apolipoprotein A-I/blood , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Coronary Thrombosis/etiology , Coronary Thrombosis/prevention & control , Coronary Vessels/metabolism , Coronary Vessels/pathology , Humans , Lipoproteins, HDL/blood , Neointima , Percutaneous Coronary Intervention/adverse effects , Prosthesis Design , Translational Research, Biomedical , Treatment Outcome
12.
Blood ; 127(5): 637-45, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26552698

ABSTRACT

The ability of von Willebrand factor (VWF) to initiate platelet adhesion depends on the number of monomers in individual VWF multimers and on the self-association of individual VWF multimers into larger structures. VWF self-association is accelerated by shear stress. We observed that VWF self-association occurs during adsorption of VWF onto surfaces, assembly of secreted VWF into hyperadhesive VWF strings on the endothelial surface, and incorporation of fluid-phase VWF into VWF fibers. VWF adsorption under static conditions increased with increased VWF purity and was prevented by a component of plasma. We identified that component as high-density lipoprotein (HDL) and its major apolipoprotein ApoA-I. HDL and ApoA-I also prevented VWF on the endothelium from self-associating into longer strands and inhibited the attachment of fluid-phase VWF onto vessel wall strands. Platelet adhesion to VWF fibers was reduced in proportion to the reduction in self-associated VWF. In a mouse model of thrombotic microangiopathy, HDL also largely prevented the thrombocytopenia induced by injection of high doses of human VWF. Finally, a potential role for ApoA-I in microvascular occlusion associated with thrombotic thrombocytopenic purpura and sepsis was revealed by the inverse relationship between the concentration of ApoA-I and that of hyperadhesive VWF. These results suggest that interference with VWF self-association would be a new approach to treating thrombotic disorders.


Subject(s)
Apolipoprotein A-I/metabolism , Lipoproteins, HDL/metabolism , Platelet Adhesiveness , Thrombosis/metabolism , von Willebrand Factor/metabolism , Animals , Apolipoprotein A-I/therapeutic use , Blood Platelets/cytology , Blood Platelets/metabolism , Humans , Lipoproteins, HDL/therapeutic use , Mice, Inbred C57BL , Protein Multimerization , Thrombocytopenia/prevention & control , von Willebrand Factor/chemistry
13.
Biochim Biophys Acta ; 1862(5): 1027-36, 2016 05.
Article in English | MEDLINE | ID: mdl-26454209

ABSTRACT

Many lines of evidence suggest a protective role for high-density lipoprotein (HDL) and its major apolipoprotein (apo)A-I in Alzheimer's Disease (AD). HDL/apoA-I particles are produced by the liver and intestine and, in addition to removing excess cholesterol from the body, are increasingly recognized to have vasoprotective functions. Here we tested the ability of reconstituted HDL (rHDL) consisting of human apoA-I reconstituted with soy phosphatidylcholine for its ability to lower amyloid beta (Aß) levels in symptomatic APP/PS1 mice, a well-characterized preclinical model of amyloidosis. Animals were treated intravenously either with four weekly doses (chronic study) or a single dose of 60mg/kg of rHDL (acute study). The major finding of our acute study is that soluble brain Aß40 and Aß42 levels were significantly reduced within 24h of a single dose of rHDL. By contrast, no changes were observed in our chronic study with respect to soluble or deposited Aß levels in animals assessed 7days after the final weekly dose of rHDL, suggesting that beneficial effects diminish as rHDL is cleared from the body. Further, rHDL-treated animals showed no change in amyloid burden, cerebrospinal fluid (CSF) Aß levels, neuroinflammation, or endothelial activation in the chronic study, suggesting that the pathology-modifying effects of rHDL may indeed be acute and may be specific to the soluble Aß pool. That systemic administration of rHDL can acutely modify brain Aß levels provides support for further investigation of the therapeutic potential of apoA-I-based agents for AD. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloidosis/therapy , Apolipoprotein A-I/therapeutic use , Brain/metabolism , Lipoproteins, HDL/therapeutic use , Peptide Fragments/metabolism , Alzheimer Disease/blood , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/blood , Amyloidosis/blood , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Apolipoprotein A-I/administration & dosage , Brain/pathology , Disease Models, Animal , Female , Humans , Lipoproteins, HDL/administration & dosage , Male , Mice , Mice, Transgenic , Peptide Fragments/blood
15.
Diabetologia ; 59(9): 1977-84, 2016 09.
Article in English | MEDLINE | ID: mdl-27193916

ABSTRACT

AIMS/HYPOTHESIS: Type 2 diabetes is characterised by decreased HDL levels, as well as the level of apolipoprotein A-I (apoA-I), the main apolipoprotein of HDLs. Pharmacological elevation of HDL and apoA-I levels is associated with improved glycaemic control in patients with type 2 diabetes. This is partly due to improved glucose uptake in skeletal muscle. METHODS: This study used kinetic modelling to investigate the impact of increasing plasma apoA-I levels on the metabolism of glucose in the db/db mouse model. RESULTS: Treatment of db/db mice with apoA-I for 2 h significantly improved both glucose tolerance (AUC 2574 ± 70 mmol/l × min vs 2927 ± 137 mmol/l × min, for apoA-I and PBS, respectively; p < 0.05) and insulin sensitivity (AUC 388.8 ± 23.8 mmol/l × min vs 194.1 ± 19.6 mmol/l × min, for apoA-I and PBS, respectively; p < 0.001). ApoA-I treatment also increased glucose uptake by skeletal muscle in both an insulin-dependent and insulin-independent manner as evidenced by increased uptake of fludeoxyglucose ([(18)F]FDG) from plasma into gastrocnemius muscle in apoA-I treated mice, both in the absence and presence of insulin. Kinetic modelling revealed an enhanced rate of insulin-mediated glucose phosphorylation (k 3) in apoA-I treated mice (3.5 ± 1.1 × 10(-2) min(-1) vs 2.3 ± 0.7 × 10(-2) min(-1), for apoA-I and PBS, respectively; p < 0.05) and an increased influx constant (3.7 ± 0.6 × 10(-3) ml min(-1) g(-1) vs 2.0 ± 0.3 × 10(-3) ml min(-1) g(-1), for apoA-I and PBS, respectively; p < 0.05). Treatment of L6 rat skeletal muscle cells with apoA-I for 2 h indicated that increased hexokinase activity mediated the increased rate of glucose phosphorylation. CONCLUSIONS/INTERPRETATION: These findings indicate that apoA-I improves glucose disposal in db/db mice by improving insulin sensitivity and enhancing glucose phosphorylation.


Subject(s)
Apolipoprotein A-I/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Fluorodeoxyglucose F18/analysis , Glucose/metabolism , Muscle, Skeletal/metabolism , Positron-Emission Tomography/methods , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , Insulin Resistance/physiology , Kinetics , Male , Mice , Muscle, Skeletal/drug effects , Phosphorylation/drug effects
16.
Stroke ; 47(1): 214-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26604250

ABSTRACT

BACKGROUND AND PURPOSE: Stroke-induced neuroinflammation and white matter damage are associated with neurological deficits. Whether D-4F, an apolipoprotein A-I mimetic peptide, treatment of stroke decreases neuroinflammation and white matter damage and improves functional outcome has not been investigated. METHODS: Adult male C57BL/6 mice were subjected to permanent middle cerebral artery occlusion (MCAo) and were orally administered saline as a vehicle control and different doses of D-4F (2, 4, 8, 16, or 32 mg/kg) starting at 2 h after MCAo and daily until euthanized at 7 days after MCAo. D-4F treatment did not alter the blood levels of high-density lipoprotein, total cholesterol, triglyceride, blood-brain barrier leakage, and infarction volume compared with control group. RESULTS: D-4F (16 mg/kg) treatment of stroke significantly improved functional outcome, increased the white matter density and the number of oligodendrocyte progenitor cells in the ischemic boundary zone of the ipsilateral striatum, and increased myelin basic protein, insulin-like growth factor-1 (IGF1), but decreased inflammatory factor Toll-like receptor-4 and tumor necrosis factor-α expression in the ischemic brain 7 days after MCAo (P<0.05, n=11/group). The neurite/axonal outgrowth in primary cultured neurons was significantly increased when treated with D-4F (100 ng/mL) and IGF1 (100 ng/mL) compared with the nontreatment control. Inhibition of IGF1 significantly attenuated D-4F or IGF1 treatment-induced axonal outgrowth. D-4F-treatment did not increase oligodendrocyte-progenitor cell proliferation but decreased oligodendrocyte-progenitor cell death. CONCLUSIONS: D-4F treatment initiated 2 h after MCAo decreases neuroinflammation and white matter damage and improves functional outcome after stroke. D-4F-induced increase in IGF1 may contribute to D-4F-induced neurite/axonal outgrowth after stroke.


Subject(s)
Apolipoprotein A-I/therapeutic use , Stroke/drug therapy , Stroke/pathology , White Matter/drug effects , White Matter/pathology , Animals , Apolipoprotein A-I/pharmacology , Cell Line, Transformed , Male , Mice , Mice, Inbred C57BL , Oligodendroglia/drug effects , Oligodendroglia/pathology
17.
J Lipid Res ; 56(4): 836-47, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25635126

ABSTRACT

This study was designed to explore the protective effect of D4F, an apoA-I mimetic peptide, on oxidized LDL (ox-LDL)-induced endoplasmic reticulum (ER) stress-CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) pathway-mediated apoptosis in macrophages. Our results showed that treating apoE knockout mice with D4F decreased the serum ox-LDL level and apoptosis in atherosclerotic lesions with concomitant downregulation of cluster of differentiation 36 (CD36) and inhibition of ER stress. In vitro, D4F inhibited macrophage-derived foam cell formation. Furthermore, like ER stress inhibitor 4-phenylbutyric acid (PBA), D4F inhibited ox-LDL- or tunicamycin (TM, an ER stress inducer)-induced reduction in cell viability and increase in lactate dehydrogenase leakage, caspase-3 activation, and apoptosis. Additionally, like PBA, D4F inhibited ox-LDL- or TM-induced activation of ER stress response as assessed by the reduced nuclear translocation of activating transcription factor 6 and the decreased phosphorylation of protein kinase-like ER kinase and eukaryotic translation initiation factor 2α, as well as the downregulation of glucose-regulated protein 78 and CHOP. Moreover, D4F mitigated ox-LDL uptake by macrophages and CD36 upregulation induced by ox-LDL or TM. These data indicate that D4F can alleviate the formation and apoptosis of macrophage-derived foam cells by suppressing CD36-mediated ox-LDL uptake and subsequent activation of the ER stress-CHOP pathway.


Subject(s)
Apolipoprotein A-I/pharmacology , Apoptosis/drug effects , CD36 Antigens/metabolism , Endoplasmic Reticulum Stress/drug effects , Foam Cells/drug effects , Gene Expression Regulation/drug effects , Transcription Factor CHOP/metabolism , Animals , Apolipoprotein A-I/therapeutic use , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Biological Transport/drug effects , Biomarkers/metabolism , Down-Regulation/drug effects , Foam Cells/cytology , Foam Cells/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/toxicity , Male , Mice , RAW 264.7 Cells , Signal Transduction/drug effects , Tunicamycin/toxicity , Up-Regulation/drug effects
19.
Am Heart J ; 169(5): 736-742.e1, 2015 May.
Article in English | MEDLINE | ID: mdl-25965722

ABSTRACT

BACKGROUND: Patients with homozygous familial hypercholesterolemia (HoFH) are at extremely elevated risk for early cardiovascular disease because of exposure to elevated low-density lipoprotein cholesterol (LDL-C) plasma levels from birth. Lowering LDL-C by statin therapy is the cornerstone for cardiovascular disease prevention, but the residual risk in HoFH remains high, emphasizing the need for additional therapies. In the present study, we evaluated the effect of serial infusions with CER-001, a recombinant human apolipoprotein A-I (apoA-I)-containing high-density lipoprotein-mimetic particle, on carotid artery wall dimensions in patients with HoFH. METHODS AND RESULTS: Twenty-three patients (mean age 39.4 ± 13.5 years, mean LDL-C 214.2 ± 81.5 mg/dL) with genetically confirmed homozygosity or compound heterozygosity for LDLR, APOB, PCSK9, or LDLRAP1 mutations received 12 biweekly infusions with CER-001 (8 mg/kg). Before and 1 hour after the first infusion, lipid values were measured. Magnetic resonance imaging (3-T magnetic resonance imaging) scans of the carotid arteries were acquired at baseline and after 24 weeks to assess changes in artery wall dimensions. After CER-001 infusion, apoA-I increased from 114.8 ± 20.7 mg/dL to 129.3 ± 23.0 mg/dL. After 24 weeks, mean vessel wall area (primary end point) decreased from 17.23 to 16.75 mm(2) (P = .008). A trend toward reduction of mean vessel wall thickness was observed (0.75 mm at baseline and 0.74 mm at follow-up, P = .0835). CONCLUSIONS: In HoFH, 12 biweekly infusions with an apoA-I-containing high-density lipoprotein-mimetic particle resulted in a significant reduction in carotid mean vessel wall area, implying that CER-001 may reverse atherogenic changes in the arterial wall on top of maximal low-density lipoprotein-lowering therapy. This finding supports further clinical evaluation of apoA-I-containing particles in patients with HoFH.


Subject(s)
Apolipoprotein A-I/pharmacology , Carotid Intima-Media Thickness , Cholesterol, LDL/blood , Coronary Artery Disease/pathology , Hyperlipoproteinemia Type II/pathology , Phospholipids/pharmacology , Recombinant Proteins/pharmacology , Adult , Apolipoprotein A-I/therapeutic use , Coronary Artery Disease/drug therapy , Female , Humans , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/genetics , Lipids/blood , Male , Middle Aged , Phospholipids/therapeutic use , Rare Diseases , Recombinant Proteins/therapeutic use
20.
Circ Res ; 113(1): e1-e9, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23613182

ABSTRACT

RATIONALE: Infusions of apolipoprotein AI (apoAI), mimetic peptides, or high-density lipoprotein (HDL) remain a promising approach for the treatment of atherosclerotic coronary disease. However, rapid clearance leads to a requirement for repeated administration of large amounts of material and limits effective plasma concentrations. OBJECTIVE: Because pegylation of purified proteins is commonly used as a method to increase their half-life in the circulation, we determined whether pegylation of apoAI or HDL would increase its plasma half-life and in turn its antiatherogenic potential. METHODS AND RESULTS: Initial pegylation attempts using lipid-poor apoAI showed a marked tendency to form multi-pegylated (PEG) species with reduced ability to promote cholesterol efflux from macrophage foam cells. However, pegylation of human holo-HDL or reconstituted phospholipid/apoAI particles (rHDL) led to selective N-terminal monopegylation of apoAI with full preservation of cholesterol efflux activity. The plasma clearance of PEG-rHDL was estimated after injection into hypercholesterolemic Apoe-/- mice; the half-life of pegylated PEG-apoAI after injection of PEG-rHDL was increased ≈7-fold compared with apoAI in nonpegylated rHDL. In comparison with nonpegylated rHDL, infusion of PEG-rHDL (40 mg/kg) into hypercholesterolemic Apoe-/- mice led to more pronounced suppression of bone marrow myeloid progenitor cell proliferation and monocytosis, as well as reduced atherosclerosis and a stable plaque phenotype. CONCLUSIONS: We describe a novel method for effective monopegylation of apoAI in HDL particles, in which lipid binding seems to protect against pegylation of key functional residues. Pegylation of apoAI in rHDL markedly increases its plasma half-life and enhances antiatherogenic properties in vivo.


Subject(s)
Aortic Diseases/prevention & control , Apolipoprotein A-I/pharmacokinetics , Atherosclerosis/prevention & control , Cholesterol/metabolism , Hypercholesterolemia/drug therapy , Lipoproteins, HDL/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Animals , Aortic Diseases/etiology , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/blood , Apolipoprotein A-I/therapeutic use , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Cell Line/drug effects , Cell Line/metabolism , Drug Evaluation, Preclinical , Foam Cells/drug effects , Foam Cells/metabolism , Half-Life , Hematopoietic Stem Cells/metabolism , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Infusions, Intravenous , Injections, Intravenous , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/blood , Lipoproteins, HDL/therapeutic use , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/prevention & control , Polyethylene Glycols/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL