Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Lipids Health Dis ; 23(1): 63, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38419057

ABSTRACT

BACKGROUND AND OBJECTIVE: Dyslipidemia is significantly more common in those with concurrent chronic kidney disease (CKD) and chronic heart failure (CHF). Sacubitril/valsartan has showcased its influence on both cardiac and renal functions, extending its influence to the modulation of lipid metabolism pathways. This study aimed to examine how sacubitril/valsartan affects lipid metabolism within the context of CKD and CHF. METHODS: This study adopted a retrospective design, focusing on a single center and involving participants who were subjected to treatment with sacubitril/valsartan and valsartan. The investigation assessed the treatment duration, with a particular emphasis on recording blood lipid indicators, including triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), apolipoprotein A (ApoA), and apolipoprotein B (ApoB). Furthermore, cardiac and renal functions, blood pressure, potassium levels, and other factors influencing the blood lipids were analyzed in both groups at identical time points. RESULTS: After 16 weeks of observation, the sacubitril/valsartan group exhibited lower TG levels compared to the valsartan group. Noteworthy was the fact that individuals undergoing sacubitril/valsartan treatment experienced an average reduction of 0.84 mmol/L in TG levels, in stark contrast to the valsartan group, which registered a decline of 0.27 mmol/L (P < 0.001). The sacubitril/valsartan group exhibited elevated levels of HDL-C and ApoA in comparison to the valsartan group (PHDL-C = 0.023, PApoA = 0.030). While TC, LDL-C, and ApoB decreased compared to baseline, the differences between groups were not statistical significance. Regarding cardiac indicators, there was an observed enhancement in the left ventricular ejection fraction (LVEF) within the sacubitril/valsartan group when compared to the baseline, and it was noticeably higher than that of the valsartan group. Spearman correlation analysis and multiple linear regression analysis revealed that medication, body mass index(BMI), and hemoglobin A1c (HbA1c) had a direct influencing effect on TG levels. CONCLUSION: Sacubitril/valsartan demonstrated improvements in lipid metabolism and cardiac indicators in patients with CKD and CHF. Specifically, it presented promising benefits in reducing TG levels. In addition, both BMI and HbA1c emerged as influential factors contributing to alterations in TG levels, independent of the administration of sacubitril/valsartan.


Subject(s)
Aminobutyrates , Heart Failure , Renal Insufficiency, Chronic , Humans , Retrospective Studies , Stroke Volume/physiology , Cholesterol, LDL , Glycated Hemoglobin , Lipid Metabolism , Tetrazoles/therapeutic use , Tetrazoles/pharmacology , Ventricular Function, Left/physiology , Valsartan/therapeutic use , Valsartan/pharmacology , Heart Failure/complications , Heart Failure/drug therapy , Biphenyl Compounds , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Drug Combinations , Apolipoproteins A/pharmacology , Apolipoproteins B , Apolipoproteins
2.
Genes (Basel) ; 11(12)2020 12 12.
Article in English | MEDLINE | ID: mdl-33322656

ABSTRACT

Estradiol (E2) enhances the anorectic action of apolipoprotein A-IV (apoA-IV), however, the intracellular mechanisms are largely unclear. Here we reported that the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway was significantly activated by E2 and apoA-IV, respectively, in primary neuronal cells isolated from rat embryonic brainstem. Importantly, the combination of E2 and apoA-IV at their subthreshold doses synergistically activated the PI3K/Akt signaling pathway. These effects, however, were significantly diminished by the pretreatment with LY294002, a selective PI3K inhibitor. E2-induced activation of the PI3K/Akt pathway was through membrane-associated ERα, because the phosphorylation of Akt was significantly increased by PPT, an ERα agonist, and by E2-BSA (E2 conjugated to bovine serum albumin) which activates estrogen receptor on the membrane. Centrally administered apoA-IV at a low dose (0.5 µg) significantly suppressed food intake and increased the phosphorylation of Akt in the nucleus tractus solitarius (NTS) of ovariectomized (OVX) rats treated with E2, but not in OVX rats treated with vehicle. These effects were blunted by pretreatment with LY294002. These results indicate that E2's regulatory role in apoA-IV's anorectic action is through the ERα-PI3K pathway in the NTS. Manipulation of the PI3K/Akt signaling activation in the NTS may provide a novel therapeutic approach for the prevention and the treatment of obesity-related disorders in females.


Subject(s)
Anorexia , Apolipoproteins A/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Solitary Nucleus , Animals , Anorexia/chemically induced , Anorexia/metabolism , Anorexia/pathology , Female , Rats , Rats, Long-Evans , Solitary Nucleus/metabolism , Solitary Nucleus/pathology
3.
Pharmacol Res ; 158: 104858, 2020 08.
Article in English | MEDLINE | ID: mdl-32430285

ABSTRACT

Angiogenesis is a finely co-ordinated, multi-step developmental process of the new vascular structure. Even though angiogenesis is regularly occurring in physiological events such as embryogenesis, in adults, it is restricted to specific tissue sites where rapid cell-turnover and membrane synthesis occurs. Both excessive and insufficient angiogenesis lead to vascular disorders such as cancer, ocular diseases, diabetic retinopathy, atherosclerosis, intra-uterine growth restriction, ischemic heart disease, stroke etc. Occurrence of altered lipid profile and vascular lipid deposition along with vascular disorders is a hallmark of impaired angiogenesis. Among lipoproteins, lipoprotein(a) needs special attention due to the presence of a multi-kringle protein subunit, apolipoprotein(a) [apo(a)], which is structurally homologous to many naturally occurring anti-angiogenic proteins such as plasminogen and angiostatin. Researchers have constructed different recombinant forms of apo(a) (rhLK68, rhLK8, RHACK2, KV-11, and AU-6) and successfully exploited its potential to inhibit unwanted angiogenesis during tumor metastasis and retinal neovascularization. Similar to naturally occurring anti-angiogenic proteins, apo(a) can directly interfere with angiogenic signaling pathways. Besides this, apo(a) can also exert its anti-angiogenic effect indirectly by inducing endothelial cell apoptosis, by inhibiting endothelial progenitor cell functions or by upregulating nuclear factors in endothelial cells via apo(a)-bound oxPLs. However, the impact of the anti-angiogenic potential of native apo(a) during physiological angiogenesis in embryos and wounded tissues is not yet explored. In this context, we review the studies so far done to demonstrate the anti-angiogenic activity of apo(a) and the recent developments in using apo(a) as a therapeutic agent to treat impaired angiogenesis during vascular disorders, with emphasis on the gaps in the literature.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apolipoproteins A/genetics , Apolipoproteins A/physiology , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/genetics , Animals , Apolipoproteins A/pharmacology , Humans
4.
Endocrinology ; 161(4)2020 04 01.
Article in English | MEDLINE | ID: mdl-32157301

ABSTRACT

Apolipoprotein A-IV (ApoA-IV) synthesized by the gut regulates lipid metabolism. Sympathetic innervation of adipose tissues also controls lipid metabolism. We hypothesized that ApoA-IV required sympathetic innervation to increase fatty acid (FA) uptake by adipose tissues and brown adipose tissue (BAT) thermogenesis. After 3 weeks feeding of either a standard chow diet or a high-fat diet (HFD), mice with unilateral denervation of adipose tissues received intraperitoneal administration of recombinant ApoA-IV protein and intravenous infusion of lipid mixture with radioactive triolein. In chow-fed mice, ApoA-IV administration increased FA uptake by intact BAT but not the contralateral denervated BAT or intact white adipose tissue (WAT). Immunoblots showed that, in chow-fed mice, ApoA-IV increased expression of lipoprotein lipase and tyrosine hydroxylase in both intact BAT and inguinal WAT (IWAT), while ApoA-IV enhanced protein levels of ß3 adrenergic receptor, adipose triglyceride lipase, and uncoupling protein 1 in the intact BAT only. In HFD-fed mice, ApoA-IV elevated FA uptake by intact epididymal WAT (EWAT) but not intact BAT or IWAT. ApoA-IV increased sympathetic activity assessed by norepinephrine turnover (NETO) rate in BAT and EWAT of chow-fed mice, whereas it elevated NETO only in EWAT of HFD-fed mice. These observations suggest that, in chow-fed mice, ApoA-IV activates sympathetic activity of BAT and increases FA uptake by BAT via innervation, while in HFD-fed mice, ApoA-IV stimulates sympathetic activity of EWAT to shunt FAs into the EWAT.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Apolipoproteins A/pharmacology , Fatty Acids/metabolism , Sympathetic Nervous System/metabolism , Thermogenesis/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Animals , Diet, High-Fat , Male , Mice , Norepinephrine/metabolism , Sympathetic Nervous System/drug effects
5.
Allergy ; 75(2): 392-402, 2020 02.
Article in English | MEDLINE | ID: mdl-31408538

ABSTRACT

BACKGROUND: Recent studies pointed to a crucial role for apolipoproteins in the pathogenesis of inflammatory diseases. However, the role of apolipoprotein-IV (ApoA-IV) in allergic inflammation has not been addressed thoroughly thus far. OBJECTIVE: Here, we explored the anti-inflammatory effects and underlying signaling pathways of ApoA-IV on eosinophil effector function in vitro and in vivo. METHODS: Migratory responsiveness, Ca2+ -flux and apoptosis of human peripheral blood eosinophils were assessed in vitro. Allergen-driven airway inflammation was assessed in a mouse model of acute house dust mite-induced asthma. ApoA-IV serum levels were determined by ELISA. RESULTS: Recombinant ApoA-IV potently inhibited eosinophil responsiveness in vitro as measured by Ca2+ -flux, shape change, integrin (CD11b) expression, and chemotaxis. The underlying molecular mechanism involved the activation of Rev-ErbA-α and induced a PI3K/PDK1/PKA-dependent signaling cascade. Systemic application of ApoA-IV prevented airway hyperresponsiveness (AHR) and airway eosinophilia in mice following allergen challenge. ApoA-IV levels were decreased in serum from allergic patients compared to healthy controls. CONCLUSION: Our data suggest that ApoA-IV is an endogenous anti-inflammatory protein that potently suppresses effector cell functions in eosinophils. Thus, exogenously applied ApoA-IV may represent a novel pharmacological approach for the treatment of allergic inflammation and other eosinophil-driven disorders.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/blood , Apolipoproteins A/administration & dosage , Apolipoproteins A/blood , Asthma/blood , Asthma/drug therapy , Rhinitis/blood , Sinusitis/blood , Adolescent , Adult , Allergens/adverse effects , Animals , Anti-Inflammatory Agents/pharmacology , Apolipoproteins A/pharmacology , Apoptosis/drug effects , Asthma/etiology , Calcium/metabolism , Cells, Cultured , Chemotaxis/drug effects , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/immunology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Pyroglyphidae/immunology , Young Adult
6.
Lipids Health Dis ; 18(1): 166, 2019 Aug 31.
Article in English | MEDLINE | ID: mdl-31470857

ABSTRACT

BACKGROUND: Atherosclerosis is an inflammatory process involving activation of monocytes recruited by various chemoattractant factors, among which lipoprotein(a) and its specific apolipoprotein apo(a). Lp(a) contains a specific apolipoprotein apo(a) which size is determined by a variable number of repeats of a specific structural domain, the kringle IV type 2 (IV-2). Lp(a) plasma concentration and apo(a) size is inversely correlated, and smaller apo(a) are major risk factors for coronary heart disease. DESIGN AND METHODS: The aim of this study was to evaluate the effect of recombinant apo(a) isoforms (containing 10, 18 or 34 kringles) on monocytes interacting with type I collagen. RESULTS: Apo(a) isoforms stimulated reactive oxygen species (ROS) and matrix metalloproteinase-9 (MMP-9) production by monocytes, and not modified monocytes adhesion on type I collagen. This effect was specific of apo(a) since no effect was observed in the presence of plasminogen and was inversely related to apo(a) size. The lysine analogue 6-aminohexanoic acid which blocks the lysine binding sites (LBS), and carboxypeptidase B (CpB) which cleaves carboxy-terminal lysine residues, abolished apo(a)-induced ROS and MMP-9 production, highlighting an effect mediated by apo(a) lysing-binding sites. CONCLUSIONS: These results indicate that activation of collagen-primed monocytes stimulated with apo(a) is a Kringle number-dependent effect and reinforce the hypothesis of a role for small size apo(a) isoforms in atherothrombosis.


Subject(s)
Apolipoproteins A/pharmacology , Collagen Type I/pharmacology , Monocytes/drug effects , Recombinant Proteins/pharmacology , Aminocaproic Acid/pharmacology , Animals , Apolipoproteins A/biosynthesis , Apolipoproteins A/chemistry , Fibronectins/pharmacology , HEK293 Cells , Humans , Matrix Metalloproteinase 9/biosynthesis , Molecular Weight , Monocytes/cytology , Monocytes/metabolism , Plasminogen/pharmacology , Primary Cell Culture , Protein Binding , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/pharmacology , Proteolysis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry
7.
Nat Commun ; 9(1): 3608, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30190457

ABSTRACT

Platelet αIIbß3 integrin and its ligands are essential for thrombosis and hemostasis, and play key roles in myocardial infarction and stroke. Here we show that apolipoprotein A-IV (apoA-IV) can be isolated from human blood plasma using platelet ß3 integrin-coated beads. Binding of apoA-IV to platelets requires activation of αIIbß3 integrin, and the direct apoA-IV-αIIbß3 interaction can be detected using a single-molecule Biomembrane Force Probe. We identify that aspartic acids 5 and 13 at the N-terminus of apoA-IV are required for binding to αIIbß3 integrin, which is additionally modulated by apoA-IV C-terminus via intra-molecular interactions. ApoA-IV inhibits platelet aggregation and postprandial platelet hyperactivity. Human apoA-IV plasma levels show a circadian rhythm that negatively correlates with platelet aggregation and cardiovascular events. Thus, we identify apoA-IV as a novel ligand of αIIbß3 integrin and an endogenous inhibitor of thrombosis, establishing a link between lipoprotein metabolism and cardiovascular diseases.


Subject(s)
Apolipoproteins A/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombosis/metabolism , Adult , Animals , Apolipoproteins A/genetics , Apolipoproteins A/pharmacology , Aspartic Acid/metabolism , Binding Sites , Circadian Rhythm/physiology , Disease Models, Animal , Humans , Mice, Inbred C57BL , Mice, Transgenic , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Postprandial Period , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Thrombosis/drug therapy
8.
Physiol Behav ; 188: 11-17, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29378187

ABSTRACT

Cholecystokinin (CCK) and apolipoprotein A-IV (ApoA-IV) are gastrointestinal peptides that play an important role in controlling energy homeostasis. Lymphatic ApoA-IV and plasma CCK secretion are mediated via a chylomicron formation-dependent pathway during a dietary lipid infusion. Given their similar roles as satiating proteins, the present study examines how the two peptides interact in their function. Specifically, this study sought to understand how ApoA-IV regulates CCK secretion. For this purpose, Cck gene expression in the small intestines of ApoA-IV knockout (ApoA-IV-KO) and wild-type (WT) mice were compared under an array of feeding conditions. When fed with a chow or high-fat diet (HFD), basal levels of Cck transcripts were significantly reduced in the duodenum of ApoA-IV-KO mice compared to WT mice. Furthermore, after an oral gavage of a lipid mixture, Cck gene expression in the duodenum was significantly reduced in ApoA-IV-KO mice relative to the change seen in WT mice. To determine the mechanism by which ApoA-IV modulates Cck gene expression, STC-1 cells were transfected with predesigned mouse lysophosphatidic acid receptor 5 (LPAR5) small interfering RNA (siRNA) to knockdown Lpar5 gene expression. In this in-vitro study, mouse recombinant ApoA-IV protein increased Cck gene expression in enteroendocrine STC-1 cells and stimulated CCK release from the STC-1 cells. However, the levels of CCK protein and Cck expression were attenuated when Lpar5 was knocked down in the STC-1 cells. Together these observations suggest that dietary lipid-induced ApoA-IV is associated with Cck synthesis in the duodenum and that ApoA-IV protein directly enhances CCK release through the activation of a LPAR5-dependent pathway.


Subject(s)
Antioxidants/pharmacology , Apolipoproteins A/pharmacology , Cholecystokinin/metabolism , Duodenum/drug effects , Gene Expression Regulation/drug effects , Animals , Apolipoproteins A/deficiency , Apolipoproteins A/genetics , Cell Line, Transformed , Cholecystokinin/genetics , Dietary Fats/administration & dosage , Dose-Response Relationship, Drug , Duodenum/metabolism , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Time Factors , Triglycerides/blood
9.
Biochem Biophys Res Commun ; 487(2): 327-332, 2017 05 27.
Article in English | MEDLINE | ID: mdl-28412351

ABSTRACT

ApoA4 exerts anti-inflammatory effects, but the mechanism remains unclear. SERPINA3 is a member of the serine proteinase inhibitor gene family, and has been shown to be involved in anti-inflammation and associated with a number of human diseases. In this study, we revealed that ApoA4 stimulates the gene expression of SERPINA3 in mouse hepatocytes both in vivo and in vitro, in a dose- and time-dependent manner. The transcriptional response of SERPINA3 to ApoA4 is regulated through the binding of ApoA4 with nuclear receptors NR4A1 and NR1D1 on the SERPINA3 promoter, which was verified with ChIP, Luciferase activity assay and RNA interference-mediated NR4A1 or NR1D1 gene knockdown. These data suggests that ApoA4 transcriptionally induced SERPINA3 expression via NR1D1 and NR4A1. Our findings may throw light on the function of ApoA4 in inflammatory responses and acute-phase reactions, as well as the development of SERPINA3 relative diseases.


Subject(s)
Apolipoproteins A/pharmacology , Hepatocytes/metabolism , Liver/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Serpins/metabolism , Animals , Hepatocytes/drug effects , Liver/drug effects , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Organ Specificity/physiology , Tissue Distribution
10.
Mol Cells ; 38(6): 573-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25997739

ABSTRACT

Apolipoprotein A-I and A-IV are protein constituents of high-density lipoproteins although their functional difference in lipoprotein metabolism is still unclear. To compare anti-atherogenic properties between apoA-I and apoA-4, we characterized both proteins in lipid-free and lipid-bound state. In lipid-free state, apoA4 showed two distinct bands, around 78 and 67 Å on native gel electrophoresis, while apoA-I showed scattered band pattern less than 71 Å. In reconstituted HDL (rHDL) state, apoA-4 showed three major bands around 101 Å and 113 Å, while apoA-I-rHDL showed almost single band around 98 Å size. Lipid-free apoA-I showed 2.9-fold higher phospholipid binding ability than apoA-4. In lipid-free state, BS3-crosslinking revealed that apoA-4 showed less multimerization tendency upto dimer, while apoA-I showed pentamerization. In rHDL state (95:1), apoA-4 was existed as dimer as like as apoA-I. With higher phospholipid content (255:1), five apoA-I and three apoA-4 were required to the bigger rHDL formation. Regardless of particle size, apoA-I-rHDL showed superior LCAT activation ability than apoA-4-rHDL. Uptake of acetylated LDL was inhibited by apoA-I in both lipid-free and lipid-bound state, while apoA-4 inhibited it only lipid-free state. ApoA-4 showed less anti-atherogenic activity with more sensitivity to glycation. In conclusion, apoA-4 showed inferior physiological functions in lipid-bound state, compared with those of apoA-I, to induce more pro-atherosclerotic properties.


Subject(s)
Apolipoprotein A-I/chemistry , Apolipoprotein A-I/physiology , Apolipoproteins A/chemistry , Apolipoproteins A/physiology , Antioxidants/chemistry , Antioxidants/physiology , Apolipoprotein A-I/isolation & purification , Apolipoprotein A-I/pharmacology , Apolipoproteins A/isolation & purification , Apolipoproteins A/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cell Line , Circular Dichroism , Humans , Lipoproteins, HDL/chemistry , Male , Phospholipids/chemistry , Structure-Activity Relationship
11.
Endocrinology ; 155(5): 1700-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24564397

ABSTRACT

Apolipoprotein AIV (Apo AIV) and cholecystokinin (CCK) are secreted in response to fat consumption, and both cause satiation via CCK 1 receptor (CCK-1R)-containing vagal afferent nerves to the nucleus of the solitary tract (NTS), where Apo AIV is also synthesized. Fasted male Long-Evans rats received ip CCK-8 or fourth-ventricular (i4vt) Apo AIV alone or in combination. Food intake and c-Fos proteins (a product of the c-Fos immediate-early gene) were assessed. i4vt Apo AIV and/or ip CCK at effective doses reduced food intake and activated c-Fos proteins in the NTS and hypothalamic arcuate nucleus and paraventricular nucleus. Blockade of the CCK-1R by i4vt lorglumide adjacent to the NTS attenuated the satiating and c-Fos-stimulating effects of CCK and Apo AIV, alone or in combination. Maintenance on a high-fat diet (HFD) for 10 weeks resulted in weight gain and attenuation of both the behavioral and c-Fos responses to a greater extent than occurred in low-fat diet-fed and pair-fed HFD animals. These observations suggest that NTS Apo AIV or/and peripheral CCK requires vagal CCK-1R signaling to elicit satiation and that maintenance on a HFD reduces the satiating capacity of these 2 signals.


Subject(s)
Apolipoproteins A/metabolism , Appetite Regulation , Cholecystokinin/metabolism , Nerve Tissue Proteins/metabolism , Neurons, Afferent/metabolism , Receptor, Cholecystokinin A/metabolism , Solitary Nucleus/metabolism , Animals , Apolipoproteins A/administration & dosage , Apolipoproteins A/genetics , Apolipoproteins A/pharmacology , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacology , Appetite Depressants/therapeutic use , Appetite Regulation/drug effects , Appetite Stimulants/administration & dosage , Appetite Stimulants/pharmacology , Appetitive Behavior/drug effects , Behavior, Animal/drug effects , Cholecystokinin/administration & dosage , Cholecystokinin/analogs & derivatives , Cholecystokinin/antagonists & inhibitors , Diet, High-Fat/adverse effects , Hormone Antagonists/administration & dosage , Hormone Antagonists/pharmacology , Infusions, Intraventricular , Injections, Intraperitoneal , Male , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Neurons, Afferent/drug effects , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Rats , Rats, Long-Evans , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Sincalide/administration & dosage , Sincalide/analogs & derivatives , Sincalide/pharmacology , Solitary Nucleus/drug effects
12.
Innate Immun ; 20(8): 867-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24323453

ABSTRACT

Endotoxemia is a major cause of chronic inflammation, and is an important pathogenic factor in the development of metabolic syndrome and atherosclerosis. Human apolipoprotein E (apoE) and apoA-I are protein components of high-density lipoprotein, which have strong anti-endotoxin activity. Here, we compared anti-endotoxin activity of Ac-hE18A-NH2 and 4F peptides, modified from model amphipathic helical 18A peptide, to mimic, respectively, apoE and apoA-I properties. Ac-hE18A-NH2, stronger than 4F, inhibited endotoxin activity and disaggregated Escherichia coli 055:B5 (wild smooth serotype). Ac-hE18A-NH2 and 4F inhibited endotoxin activity of E. coli 026:B6 (rough-like serotype) to a similar degree. This suggests that Ac-hE18A-NH2 as a dual-domain molecule might interact with both the lipid A and headgroup of smooth LPS, whereas 4F binds lipid A. In C57BL/6 mice, Ac-hE18A-NH2 was superior to 4F in inhibiting the inflammatory responses mediated by E. coli 055:B5, but not E. coli 026:B6. However, in THP-1 cells, isolated human primary leukocytes, and whole human blood, Ac-hE18A-NH2 reduced responses more strongly than 4F to both E. coli serotypes either when peptides were pre-incubated or co-incubated with LPS, indicating that Ac-hE18A-NH2 also has strong anti-inflammatory effects independent of endotoxin-neutralizing properties. In conclusion, Ac-hE18A-NH2 is more effective than 4F in inhibiting LPS-mediated inflammation, which opens prospective clinical applications for Ac-hE18A-NH2.


Subject(s)
Apolipoproteins A/pharmacology , Apolipoproteins E/pharmacology , Endotoxins/antagonists & inhibitors , Peptides/pharmacology , Amino Acid Sequence , Animals , Biomimetics , Cell Line , Female , Humans , In Vitro Techniques , Leukocytes/drug effects , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Molecular Sequence Data
13.
Physiol Behav ; 120: 136-42, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911688

ABSTRACT

Rodent apoA-IV is expressed predominantly in small intestine and also expressed to a small extent in liver and hypothalamus. ApoA-IV has been shown to inhibit food intake in rats when injected centrally. In the current study, we hypothesize that a specific sequence within rat apoA-IV is responsible for mediating the anorectic effect. We use a bacterial expression system to generate truncation mutants (Δ249-371, Δ117-371 and Δ1-61) of rat apoA-IV and assess the ability of various regions of the molecule to inhibit food intake. The results indicate that a responsible sequence exists within the N-terminal 61 amino acids of rat apoA-IV. Synthetic peptides (1-30 EVTSDQVANVMWDYFTQLSNNAKEAVEQLQ, 1-15 EVTSDQVANVMWDYF and 17-30 QLSNNAKEAVEQLQ) were used to specify the region in between residues 1 and 30. A 14-mer peptide (17-30) encompassing this sequence was capable of reducing food intake in a dose-dependent manner whereas a peptide designed on a more C-terminal region (211-232) of apoA-IV (QEKLNHQMEGLAFQMKKNAEEL) failed to exhibit the dose-dependent anorectic effect. The isolation of this sequence provides a valuable tool for future work directed at identifying apoA-IV binding proteins and is a key step for exploring the potential of therapeutic manipulation of food intake via this pathway.


Subject(s)
Apolipoproteins A/chemistry , Apolipoproteins A/pharmacology , Appetite Depressants , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Eating/drug effects , Lipopolysaccharides/pharmacology , Male , Molecular Sequence Data , Mutagenesis , Peptides/chemical synthesis , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Structure-Activity Relationship
14.
J Lipid Res ; 54(9): 2341-53, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23828780

ABSTRACT

MDCO-216, a complex of dimeric recombinant apoA-IMilano (apoA-IM) and palmitoyl-oleoyl-phosphatidylcholine (POPC), was administered to cynomolgus monkeys at 30, 100, and 300 mg/kg every other day for a total of 21 infusions, and effects on lipids, (apo)lipoproteins, and ex-vivo cholesterol efflux capacity were monitored. After 7 or 20 infusions, free cholesterol (FC) and phospholipids (PL) were strongly increased, and HDL-cholesterol (HDL-C), apoA-I, and apoA-II were strongly decreased. We then measured short-term effects on apoA-IM, lipids, and (apo)lipoproteins after the first or the last infusion. After the first infusion, PL and FC went up in the HDL region and also in the LDL and VLDL regions. ApoE shifted from HDL to LDL and VLDL regions, while ApoA-IM remained located in the HDL region. On day 41, ApoE levels were 8-fold higher than on day 1, and FC, PL, and apoE resided mostly in LDL and VLDL regions. Drug infusion quickly decreased the endogenous cholesterol esterification rate. ABCA1-mediated cholesterol efflux on day 41 was markedly increased, whereas scavenger receptor type B1 (SRB1) and ABCG1-mediated effluxes were only weakly increased. Strong increase of FC is due to sustained stimulation of ABCA1-mediated efflux, and drop in HDL and formation of large apoE-rich particles are due to lack of LCAT activation.


Subject(s)
Apolipoprotein A-I/administration & dosage , Apolipoproteins A/administration & dosage , Apolipoproteins A/pharmacology , Cholesterol/blood , Phosphatidylcholines/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Animals , Apolipoproteins A/blood , Apolipoproteins E/blood , Biological Transport/drug effects , Cholesterol/metabolism , Drug Combinations , Esterification/drug effects , Female , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Macaca fascicularis , Male , Recombinant Fusion Proteins/blood , Time Factors
15.
Protein Eng Des Sel ; 26(6): 425-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23571426

ABSTRACT

Angiogenesis is crucial for tumor growth and metastasis. Blocking this process is, therefore, a potentially powerful approach for the treatment of cancer. Human apolipoprotein(a) kringle V (rhLK8) is an angiogenesis inhibitor and is currently under development as an anti-cancer therapeutic. However, a relatively short in vivo half-life limits its widespread clinical use. This study was performed to evaluate whether fusion of an Fc domain to rhLK8 can extend plasma half-life. RhLK8-Fc fusion protein was expressed in CHO DG44 cells as a dimer and was readily purified by protein G affinity chromatography. The anti-angiogenic activity of rhLK8-Fc was similar to that of rhLK8, as determined by migration and tube formation assays with endothelial cells in vitro and a chorioallantoic membrane assay in vivo. Pharmacokinetic profiles in mice after single intravenous administration of rhLK8 or rhLK8-Fc showed that Fc fusion significantly increased the elimination half-life (t(½)) and the systemic exposure (AUC(inf)) of the protein, in parallel with a significant decrease in total clearance (CL). These data suggest that Fc fusion to rhLK8 is a powerful strategy for extending the plasma half-life of rhLK8 without affecting its anti-angiogenic activity, and could thus improve the clinical applicability of rhLK8.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Apolipoproteins A/pharmacokinetics , Immunoglobulin Fc Fragments/blood , Peptide Fragments/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Apolipoproteins A/chemistry , Apolipoproteins A/pharmacology , CHO Cells , Cell Movement/drug effects , Cricetinae , Cricetulus , Half-Life , Human Umbilical Vein Endothelial Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/pharmacology , Male , Mice , Mice, Inbred ICR , Neovascularization, Physiologic/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
16.
PLoS One ; 8(1): e52287, 2013.
Article in English | MEDLINE | ID: mdl-23326327

ABSTRACT

Elevated plasma concentrations of lipoprotein(a) are associated with increased risk for atherothrombotic diseases. Apolipoprotein(a), the unique glycoprotein component of lipoprotein(a), is characterized by the presence of multiple kringle domains, and shares a high degree of sequence homology with the serine protease zymogen plasminogen. It has been shown that angiostatin, a proteolytic fragment of plasminogen containing kringles 1-4, can effectively inhibit angiogenesis. Moreover, proteolytic fragments of plasminogen containing kringle 5 are even more potent inhibitors of angiogenesis than angiostatin. Despite its strong similarity with plasminogen, the role of apolipoprotein(a) in angiogenesis remains controversial, with both pro- and anti-angiogenic effects reported. In the current study, we evaluated the ability of apolipoprotein(a) to inhibit VEGF- and angiopoietin-induced tube formation in human umbilical cord endothelial cells. A 17 kringle-containing form of recombinant apo(a) (17K), corresponding to a well-characterized, physiologically-relevant form of the molecule, effectively inhibited tube formation induced by either VEGF or angiopoietin-1. Using additional recombinant apolipoprotein(a) (r-apo(a)) variants, we demonstrated that this effect was dependent on the presence of an intact lysine-binding site in kringle V domain of apo(a), but not on the presence of the functional lysine-binding site in apo(a) kringle IV type 10; sequences within in the amino-terminal half of the molecule were also not required for the inhibitory effects of apo(a). We also showed that the apo(a)-mediated inhibition tube formation could be reversed, in part by the addition of plasmin or urokinase plasminogen activator, or by removal of plasminogen from the system. Further, we demonstrated that apo(a) treated with glycosidases to remove sialic acid was significantly less effective in inhibiting tube formation. This is the first report of a functional role for the glycosylation of apo(a) although the mechanisms underlying this observation remain to be determined in the context of angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apolipoproteins A/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Angiopoietin-1/pharmacology , Apolipoproteins A/genetics , Apolipoproteins A/metabolism , Cell Line , Fibrinolysin/pharmacology , Glycosylation , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Kringles/genetics , Recombinant Proteins/pharmacology , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
17.
J Pharmacol Exp Ther ; 344(1): 50-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23042953

ABSTRACT

The bihelical apolipoprotein mimetic peptide 5A effluxes cholesterol from cells and reduces inflammation and atherosclerosis in animal models. We investigated how hydrophobic residues in the hinge region between the two helices are important in the structure and function of this peptide. By simulated annealing analysis and molecular dynamics modeling, two hydrophobic amino acids, F-18 and W-21, in the hinge region were predicted to be relatively surface-exposed and to interact with the aqueous solvent. Using a series of 5A peptide analogs in which F-18 or W-21 was changed to either F, W, A, or E, only peptides with hydrophobic amino acids in these two positions were able to readily bind and solubilize phospholipid vesicles. Compared with active peptides containing F or W, peptides containing E in either of these two positions were more than 10-fold less effective in effluxing cholesterol by the ABCA1 transporter. Intravenous injection of 5A in C57BL/6 mice increased plasma-free cholesterol (5A: 89.9 ± 13.6 mg/dl; control: 38.7 ± 4.3 mg/dl (mean ± S.D.); P < 0.05) and triglycerides (5A: 887.0 ± 172.0 mg/dl; control: 108.9 ± 9.9 mg/dl; P < 0.05), whereas the EE peptide containing E in both positions had no effect. Finally, 5A increased cholesterol efflux approximately 2.5-fold in vivo from radiolabeled macrophages, whereas the EE peptide was inactive. These results provide a rationale for future design of therapeutic apolipoprotein mimetic peptides and provide new insights into the interaction of hydrophobic residues on apolipoproteins with phospholipids in the lipid microdomain created by the ABCA1 transporter during the cholesterol efflux process.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Amino Acids/chemistry , Apolipoproteins A/pharmacology , Cholesterol/metabolism , ATP Binding Cassette Transporter 1 , Amino Acid Sequence , Animals , Cell Line , Circular Dichroism , Cricetinae , Female , Lipids/blood , Lipoproteins/blood , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Phospholipids/metabolism
18.
Neoplasia ; 14(4): 335-43, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22577348

ABSTRACT

Antivascular therapy has emerged as a rational strategy to improve the treatment of androgen-independent prostate cancer owing to the necessity of establishing a vascular network for the growth and progression of the primary and metastatic tumor. We determined whether recombinant human apolipoprotein(a) kringle V, rhLK8, produces therapeutic efficacy in an orthotopic human prostate cancer animal model. Fifty thousand androgen-independent human prostate cancer cells (PC-3MM2) were injected into the prostate of nude mice. After 3 days, these mice were randomized to receive the vehicle solution (intraperitoneally [i.p.], daily), paclitaxel (8 mg/kg i.p., weekly), rhLK8 (50 mg/kg i.p., daily), or a combination of paclitaxel and rhLK8 for 4 weeks. Treatment with paclitaxel or rhLK8 alone did not show significant therapeutic effects on tumor incidence or on tumor size compared with the control group. The combination of rhLK8 and paclitaxel significantly reduced tumor size and incidence of lymph node metastasis. Significant reduction in microvessel density and cellular proliferation and induction of apoptosis of tumor cells, and tumor-associated endothelial cells, were also achieved. Similarly, PC-3MM2 tumors growing in the tibia showed significant suppression of tumor growth and lymph node metastasis by the combination treatment with rhLK8 and paclitaxel. The integrity of the bone was significantly preserved, and apoptosis of tumor cells and tumor-associated endothelial cells was increased. In conclusion, these results suggest that targeting the tumor microenvironment with the antivascular effect of rhLK8 combined with conventional cytotoxic chemotherapy could be a new and effective approach in the treatment of androgen-independent prostate cancer and their metastases.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apolipoproteins A/pharmacology , Kringles , Peptide Fragments/pharmacology , Prostatic Neoplasms/pathology , Animals , Apolipoproteins A/chemistry , Disease Models, Animal , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Paclitaxel/pharmacology , Prostatic Neoplasms/blood supply , Recombinant Proteins/pharmacology , Xenograft Model Antitumor Assays
19.
Diabetes ; 61(6): 1599-608, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22427380

ABSTRACT

Retinal neovascularization is observed in progression of diabetic retinopathy. New vessels grow into the vitreous cavity in proliferative diabetic retinopathy, resulting in traction retinal detachment and vitreous hemorrhage. To overcome the catastrophic visual loss due to these complications, efforts have been focused on the treatment of retinal neovascularization. In this study, we demonstrated the inhibitory effect of recombinant human apolipoprotein(a) kringle V (rhLK8) in an animal model of ischemia-induced retinal neovascularization. rhLK8 induced no definite toxicity on endothelial cells and retinal tissues at the therapeutic dosage. Interestingly, rhLK8 showed antiangiogenic effect, particularly on fibronectin-mediated migration of endothelial cells. Further experiments demonstrated high binding affinity of rhLK8 to α3ß1 integrin, and suppression of it might be the mechanism of antiangiogenic effect of rhLK8. Furthermore, rhLK8 inhibited phosphorylation of focal adhesion kinase, resulting in suppression of activation of consequent p130CAS-Jun NH(2)-terminal kinase. Taken together, our data suggested the possible application of rhLK8 in the treatment of retinal neovascularization by suppression of fibronectin-mediated angiogenesis.


Subject(s)
Apolipoproteins A/therapeutic use , Endothelial Cells/drug effects , Fibronectins/metabolism , Retina/drug effects , Retinal Neovascularization/drug therapy , Animals , Apolipoproteins A/pharmacology , Cell Movement/drug effects , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Phosphorylation/drug effects , Retina/metabolism , Retinal Neovascularization/metabolism
20.
Biol Chem ; 392(4): 347-56, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21194375

ABSTRACT

Many proteins in the fibrinolysis pathway contain antiangiogenic kringle domains. Owing to the high degree of homology between kringle domains, there has been a safety concern that antiangiogenic kringles could interact with common kringle proteins during fibrinolysis leading to adverse effects in vivo. To address this issue, we investigated the effects of several antiangiogenic kringle proteins including angiostatin, apolipoprotein(a) kringles IV(9)-IV(10)-V (LK68), apolipoprotein(a) kringle V (rhLK8) and a derivative of rhLK8 mutated to produce a functional lysine-binding site (Lys-rhLK8) on the entire fibrinolytic process in vitro and analyzed the role of lysine binding. Angiostatin, LK68 and Lys-rhLK8 increased clot lysis time in a dose-dependent manner, inhibited tissue-type plasminogen activator-mediated plasminogen activation on a thrombin-modified fibrinogen (TMF) surface, showed binding to TMF and significantly decreased the amount of plasminogen bound to TMF. The inhibition of fibrinolysis by these proteins appears to be dependent on their functional lysine-binding sites. However, rhLK8 had no effect on these processes owing to an inability to bind lysine. Collectively, these results indicate that antiangiogenic kringles without lysine binding sites might be safer with respect to physiological fibrinolysis than lysine-binding antiangiogenic kringles. However, the clinical significance of these findings will require further validation in vivo.


Subject(s)
Apolipoproteins A/chemistry , Apolipoproteins A/pharmacology , Fibrinolysis/drug effects , Kringles , Lysine , Plasminogen/chemistry , Plasminogen/pharmacology , Amino Acid Sequence , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Angiostatins/adverse effects , Angiostatins/chemistry , Angiostatins/metabolism , Angiostatins/pharmacology , Apolipoproteins A/adverse effects , Apolipoproteins A/metabolism , Binding Sites , Dose-Response Relationship, Drug , Fibrin/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Plasminogen/adverse effects , Plasminogen/metabolism , Thrombin/chemistry , Thrombin/metabolism , Tissue Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...