Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513710

ABSTRACT

Stimulation of thermogenesis in brown adipose tissue (BAT) could have far-reaching health benefits in combatting obesity and obesity-related complications. Apolipoprotein A-IV (ApoA-IV), produced by the gut and the brain in the presence of dietary lipids, is a well-known short-term satiating protein. While our previous studies have demonstrated reduced diet-induced thermogenesis in ApoA-IV-deficient mice, it is unclear whether this reduction is due to a loss of peripheral or central effects of ApoA-IV. We hypothesized that central administration of ApoA-IV stimulates BAT thermogenesis and that sympathetic and sensory innervation is necessary for this action. To test this hypothesis, mice with unilateral denervation of interscapular BAT received central injections of recombinant ApoA-IV protein or artificial cerebrospinal fluid (CSF). The effects of central ApoA-IV on BAT temperature and thermogenesis in mice with unilateral denervation of the intrascapular BAT were monitored using transponder probe implantation, qPCR, and immunoblots. Relative to CSF, central administration of ApoA-IV significantly increased temperature and UCP expression in BAT. However, all of these effects were significantly attenuated or prevented in mice with unilateral denervation. Together, these results clearly demonstrate that ApoA-IV regulates BAT thermogenesis centrally, and this effect is mediated through sympathetic and sensory nerves.


Subject(s)
Adipose Tissue, Brown/physiology , Apolipoproteins A/administration & dosage , Gene Expression Regulation/drug effects , Sympathetic Nervous System/physiology , Thermogenesis/drug effects , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/metabolism , Animals , Apolipoproteins A/deficiency , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Gene Expression Regulation/genetics , Lipase/genetics , Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins , Third Ventricle/physiology , Tyrosine 3-Monooxygenase/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
2.
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
3.
Biochem Biophys Res Commun ; 494(1-2): 152-157, 2017 12 09.
Article in English | MEDLINE | ID: mdl-29037812

ABSTRACT

Apolipoprotein A-IV (apoA-IV) is a satiation factor that acts in the hypothalamus, however, the intracellular mechanisms responsible for this action are still largely unknown. Here we report that apoA-IV treatment elicited a rapid activation of the phosphatidylinositol-3-kinase (PI3K) signaling pathway in cultured primary hypothalamic neurons, and this effect was significantly attenuated by pretreatment with LY294002, an inhibitor of the PI3K pathway. To determine if the activation of PI3K is required for apoA-IV's inhibitory effect on food intake, apoA-IV was administered intracerebroventricularly. We found that apoA-IV significantly reduced food intake and activated PI3K signaling in the hypothalamus, and these effects were abolished by icv pre-treatment with LY294002. To identify the distinct brain sites where apoA-IV exerts its anorectic action, apoA-IV was administered into the ventromedial hypothalamus (VMH) through implanted bilateral cannula. At a low dose (0.5 µg), apoA-IV significantly inhibited food intake and activated PI3K signaling pathway in the VMH of lean rats, but not in high-fat diet-induced obese (DIO) rats. These results collectively demonstrate a critical role of the PI3K/Akt pathway in apoA-IV's anorectic action in lean rats and suggest a defective PI3K pathway in the VMH is responsible for the impaired apoA-IV's anorectic action in the DIO animals.


Subject(s)
Apolipoproteins A/metabolism , Appetite Depressants/metabolism , Hypothalamus/metabolism , Animals , Apolipoproteins A/administration & dosage , Appetite Depressants/administration & dosage , Cells, Cultured , Diet, High-Fat/adverse effects , Eating/drug effects , Female , Hypothalamus/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Obesity/drug therapy , Obesity/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Long-Evans , Signal Transduction/drug effects
4.
Biomaterials ; 112: 218-233, 2017 01.
Article in English | MEDLINE | ID: mdl-27768975

ABSTRACT

Anti-vascular endothelial growth factor (VEGF) therapies are widely used for the treatment of neovascular fundus diseases such as diabetic retinopathy. However, these agents need to be injected intravitreally, because their strong hydrophilicity and high molecular weight prevent them from penetrating cell membranes and complex tissue barriers. Moreover, the repeated injections that are required can cause infection and tissue injury. In this study, we used in vivo-directed evolution phage display technology to identify a novel dodecapeptide, named CC12, with the ability to penetrate the ocular barrier in a noninvasive (via conjunctival sac instillation) or minimally invasive (via retrobulbar injection) manner. KV11, an antiangiogenesis peptide previously demonstrated to inhibit pathological neovascularization in the retina, was then used as a model antiangiogenesis cargo for CC12. We found that conjugation of KV11 peptide with CC12 peptide facilitated the delivery of KV11 to the retina, resulting in significant inhibition of retinal neovascularization development via topical application without tissue toxicity. Collectively, our data of multilevel evaluations demonstrate that CC12 may enable the noninvasive to minimally invasive intraocular delivery of antiangiogenic therapeutics.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Apolipoproteins A/pharmacokinetics , Directed Molecular Evolution/methods , Oligopeptides/administration & dosage , Peptide Fragments/pharmacokinetics , Retinal Neovascularization/drug therapy , Administration, Ophthalmic , Angiogenesis Inhibitors/genetics , Animals , Animals, Newborn , Apolipoproteins A/administration & dosage , Injections, Intraocular/methods , Male , Mice , Mice, Inbred C57BL , Oligopeptides/chemistry , Oligopeptides/genetics , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Treatment Outcome
5.
Lancet ; 388(10057): 2239-2253, 2016 11 05.
Article in English | MEDLINE | ID: mdl-27665230

ABSTRACT

BACKGROUND: Elevated lipoprotein(a) (Lp[a]) is a highly prevalent (around 20% of people) genetic risk factor for cardiovascular disease and calcific aortic valve stenosis, but no approved specific therapy exists to substantially lower Lp(a) concentrations. We aimed to assess the efficacy, safety, and tolerability of two unique antisense oligonucleotides designed to lower Lp(a) concentrations. METHODS: We did two randomised, double-blind, placebo-controlled trials. In a phase 2 trial (done in 13 study centres in Canada, the Netherlands, Germany, Denmark, and the UK), we assessed the effect of IONIS-APO(a)Rx, an oligonucleotide targeting apolipoprotein(a). Participants with elevated Lp(a) concentrations (125-437 nmol/L in cohort A; ≥438 nmol/L in cohort B) were randomly assigned (in a 1:1 ratio in cohort A and in a 4:1 ratio in cohort B) with an interactive response system to escalating-dose subcutaneous IONIS-APO(a)Rx (100 mg, 200 mg, and then 300 mg, once a week for 4 weeks each) or injections of saline placebo, once a week, for 12 weeks. Primary endpoints were mean percentage change in fasting plasma Lp(a) concentration at day 85 or 99 in the per-protocol population (participants who received more than six doses of study drug) and safety and tolerability in the safety population. In a phase 1/2a first-in-man trial, we assessed the effect of IONIS-APO(a)-LRx, a ligand-conjugated antisense oligonucleotide designed to be highly and selectively taken up by hepatocytes, at the BioPharma Services phase 1 unit (Toronto, ON, Canada). Healthy volunteers (Lp[a] ≥75 nmol/L) were randomly assigned to receive a single dose of 10-120 mg IONIS-APO(a)LRx subcutaneously in an ascending-dose design or placebo (in a 3:1 ratio; single-ascending-dose phase), or multiple doses of 10 mg, 20 mg, or 40 mg IONIS-APO(a)LRx subcutaneously in an ascending-dose design or placebo (in an 8:2 ratio) at day 1, 3, 5, 8, 15, and 22 (multiple-ascending-dose phase). Primary endpoints were mean percentage change in fasting plasma Lp(a) concentration, safety, and tolerability at day 30 in the single-ascending-dose phase and day 36 in the multiple-ascending-dose phase in participants who were randomised and received at least one dose of study drug. In both trials, the randomised allocation sequence was generated by Ionis Biometrics or external vendor with a permuted-block randomisation method. Participants, investigators, sponsor personnel, and clinical research organisation staff who analysed the data were all masked to the treatment assignments. Both trials are registered with ClinicalTrials.gov, numbers NCT02160899 and NCT02414594. FINDINGS: From June 25, 2014, to Nov 18, 2015, we enrolled 64 participants to the phase 2 trial (51 in cohort A and 13 in cohort B). 35 were randomly assigned to IONIS-APO(a)Rx and 29 to placebo. At day 85/99, participants assigned to IONIS-APO(a)Rx had mean Lp(a) reductions of 66·8% (SD 20·6) in cohort A and 71·6% (13·0) in cohort B (both p<0·0001 vs pooled placebo). From April 15, 2015, to Jan 11, 2016, we enrolled 58 healthy volunteers to the phase 1/2a trial of IONIS-APO(a)-LRx. Of 28 participants in the single-ascending-dose phase, three were randomly assigned to 10 mg, three to 20 mg, three to 40 mg, six to 80 mg, six to 120 mg, and seven to placebo. Of 30 participants in the multiple-ascending-dose phase, eight were randomly assigned to 10 mg, eight to 20 mg, eight to 40 mg, and six to placebo. Significant dose-dependent reductions in mean Lp(a) concentrations were noted in all single-dose IONIS-APO(a)-LRx groups at day 30. In the multidose groups, IONIS-APO(a)-LRx resulted in mean reductions in Lp(a) of 66% (SD 21·8) in the 10 mg group, 80% (SD 13·7%) in the 20 mg group, and 92% (6·5) in the 40 mg group (p=0·0007 for all vs placebo) at day 36. Both antisense oligonucleotides were safe. There were two serious adverse events (myocardial infarctions) in the IONIS-APO(a)Rx phase 2 trial, one in the IONIS-APO(a)Rx and one in the placebo group, but neither were thought to be treatment related. 12% of injections with IONIS-APO(a)Rx were associated with injection-site reactions. IONIS-APO(a)-LRx was associated with no injection-site reactions. INTERPRETATION: IONIS-APO(a)-LRx is a novel, tolerable, potent therapy to reduce Lp(a) concentrations. IONIS-APO(a)-LRx might mitigate Lp(a)-mediated cardiovascular risk and is being developed for patients with elevated Lp(a) concentrations with existing cardiovascular disease or calcific aortic valve stenosis. FUNDING: Ionis Pharmaceuticals.


Subject(s)
Apolipoproteins A/administration & dosage , Apoprotein(a)/antagonists & inhibitors , Lipoprotein(a) , Oligonucleotides, Antisense/administration & dosage , Apolipoproteins A/genetics , Cardiovascular Diseases/drug therapy , Double-Blind Method , Female , Humans , Lipoprotein(a)/blood , Lipoprotein(a)/genetics , Male , Middle Aged , Risk Factors , Treatment Outcome
7.
Lancet ; 386(10002): 1472-83, 2015 Oct 10.
Article in English | MEDLINE | ID: mdl-26210642

ABSTRACT

BACKGROUND: Lipoprotein(a) (Lp[a]) is a risk factor for cardiovascular disease and calcific aortic valve stenosis. No effective therapies to lower plasma Lp(a) concentrations exist. We have assessed the safety, pharmacokinetics, and pharmacodynamics of ISIS-APO(a)Rx, a second-generation antisense drug designed to reduce the synthesis of apolipoprotein(a) (apo[a]) in the liver. METHODS: In this randomised, double-blind, placebo-controlled, phase 1 study at the PAREXEL Clinical Pharmacology Research Unit (Harrow, Middlesex, UK), we screened for healthy adults aged 18-65 years, with a body-mass index less than 32·0 kg/m(2), and Lp(a) concentration of 25 nmol/L (100 mg/L) or more. Via a randomisation technique, we randomly assigned participants to receive a single subcutaneous injection of ISIS-APO(a)Rx (50 mg, 100 mg, 200 mg, or 400 mg) or placebo (3:1) in the single-dose part of the study or to receive six subcutaneous injections of ISIS-APO(a)Rx (100 mg, 200 mg, or 300 mg, for a total dose exposure of 600 mg, 1200 mg, or 1800 mg) or placebo (4:1) during a 4 week period in the multi-dose part of the study. Participants, investigators, and study staff were masked to the treatment assignment, except for the pharmacist who prepared the ISIS-APO(a)Rx or placebo. The primary efficacy endpoint was the percentage change from baseline in Lp(a) concentration at 30 days in the single-dose cohorts and at 36 days for the multi-dose cohorts. Safety and tolerability was assessed 1 week after last dose and included determination of the incidence, severity, and dose relation of adverse events and changes in laboratory variables, including lipid panel, routine haematology, blood chemistry, urinalysis, coagulation, and complement variables. Other assessments included vital signs, a physical examination, and 12-lead electrocardiograph. This trial is registered with European Clinical Trials Database, number 2012-004909-27. FINDINGS: Between Feb 27, 2013, and July 15, 2013, 47 (23%) of 206 screened volunteers were randomly assigned to receive ISIS-APO(a)Rx as a single-dose or multi-dose of ascending concentrations or placebo. In the single-dose study, we assigned three participants to receive 50 mg ISIS-APO(a)Rx, three participants to receive 100 mg ISIS-APO(a)Rx, three participants to receive 200 mg ISIS-APO(a)Rx, three participants to receive 400 mg ISIS-APO(a)Rx, and four participants to receive placebo. All 16 participants completed treatment and follow-up and were included in the pharmacodynamics, pharmacokinetics, and safety analyses. For the multi-dose study, we assigned eight participants to receive six doses of 100 mg ISIS-APO(a)Rx, nine participants to receive six doses of 200 mg ISIS-APO(a)Rx, eight participants to receive six doses of 300 mg ISIS-APO(a)Rx, and six participants to receive six doses of placebo. Whereas single doses of ISIS-APO(a)Rx (50-400 mg) did not decrease Lp(a) concentrations at day 30, six doses of ISIS-APO(a)Rx (100-300 mg) resulted in dose-dependent, mean percentage decreases in plasma Lp(a) concentration of 39·6% from baseline in the 100 mg group (p=0·005), 59·0% in the 200 mg group (p=0·001), and 77·8% in the 300 mg group (p=0·001). Similar reductions were observed in the amount of oxidized phospholipids associated with apolipoprotein B-100 and apolipoprotein(a). Mild injection site reactions were the most common adverse events. INTERPRETATION: ISIS-APO(a)Rx results in potent, dose-dependent, selective reductions of plasma Lp(a). The safety and tolerability support continued clinical development of ISIS-APO(a)Rx as a potential therapeutic drug to reduce the risk of cardiovascular disease and calcific aortic valve stenosis in patients with elevated Lp(a) concentration. FUNDING: Isis Pharmaceuticals.


Subject(s)
Apolipoproteins A/administration & dosage , Apoprotein(a)/antagonists & inhibitors , Oligonucleotides, Antisense/administration & dosage , Adolescent , Adult , Aged , Apolipoproteins A/genetics , Double-Blind Method , Healthy Volunteers , Humans , Middle Aged , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacokinetics , Oligonucleotides/pharmacology , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/pharmacology , Protein Binding , RNA, Messenger/metabolism , Treatment Outcome , Young Adult
9.
J Cardiovasc Pharmacol Ther ; 20(1): 66-75, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24742767

ABSTRACT

Apolipoprotein A-IMilano (ApoA-IM), a naturally occurring Arg173 to Cys mutant of ApoA-I, has been shown to reduce atherosclerosis in animal models and in a small phase 2 human trial. We have shown superior atheroprotective effects of ApoA-IM gene compared with wild-type ApoA-I gene using transplantation of retrovirally transduced bone marrow in ApoA-I/ApoE null mice. In this study, we compared the antiatherogenic efficacy of ApoA-IM gene transfer using Recombinant adeno-associated virus (rAAV) 2 or rAAV8 as vectors in ApoA-I/ApoE null mice. Mice received a single intravenous injection of 1.2 × 10(12) vector genomes of AAV2 or AAV8 vectors expressing ApoA-IM or control empty vectors (12 mice/group). Circulating levels of ApoA-IM were higher in recipients of AAV8 compared with AAV2 at 4, 12, and 20 weeks postinjection. Qualitative polymerase chain reaction analysis of RNA collected from different tissues showed that the AAV8-mediated gene transfer resulted in a more efficient transgene expression in the heart, brain, liver, lung, spleen, and kidney of the recipient mice compared with AAV2. Intravenous AAV8-ApoA-IM injection reduced atherosclerosis in the whole aorta (P < .01), aortic sinuses (P < .05), and brachiocephalic arteries (P < .05) compared with the vector control, whereas there was no statistically significant reduction in atherosclerosis in mice receiving intravenous AAV2-ApoA-IM. The ApoA-IM gene was expressed in the aortic tissue of mice receiving AAV8 ApoA-IM but not in those receiving AAV2 ApoA-IM. Immunostaining showed that compared with the vector control, there was reduced macrophage content in the brachiocephalic (P < .05) and aortic sinus plaques (P < .05) of AAV8 ApoA-IM recipients but not in the recipients of AAV2 ApoA-IM. Thus, intravenous injection of AAV8 is more effective than intravenous injection of AAV2 in the expression of ApoA-IM gene. These data provide support for the potential feasibility of this approach for atheroprotection in humans.


Subject(s)
Apolipoproteins A/genetics , Atherosclerosis/genetics , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Hypercholesterolemia/genetics , Administration, Intravenous , Animals , Apolipoproteins A/administration & dosage , Atherosclerosis/therapy , Female , Genetic Vectors/administration & dosage , Hypercholesterolemia/therapy , Male , Mice , Mice, Knockout
10.
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
11.
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
12.
Toxicol Lett ; 212(3): 298-306, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22659101

ABSTRACT

Because anti-angiogenic agents have shown various toxicities in clinical applications, the determination of their toxicities and their reversibility is important in the design of clinical trials. This study was performed to investigate the potential toxicities of an angiogenesis inhibitor, apolipoprotein(a) (Apo(a)) kringle V (rhLK8) in rats. Rats administered an intravenous (IV) bolus injection of rhLK8 (200 mg/kg) for 7 days showed significant increases in serum blood urea nitrogen (BUN), creatinine and the BUN/creatinine ratio, which was compatible with acute tubulointerstitial nephritis (TIN) in pathological examination. Because anti-angiogenic therapies are usually based on long-term treatment strategies, rats were administered 200 mg/kg/day of rhLK8 by intravenous infusion for 28 days. Rats receiving 200 mg/kg of rhLK8 showed abnormal serological and histologic findings, but their levels returned to within normal ranges 2 weeks after the cessation of administration. The creatinine clearance rate (CCr) was not affected by rhLK8 treatment. Collectively, our data indicate that the intravenous infusion of rhLK8 at therapeutic doses may induce renal toxicities, such as acute TIN, but these toxicities are clinically tolerable and reversible with close monitoring and a recovery period.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/toxicity , Apolipoproteins A/administration & dosage , Apolipoproteins A/toxicity , Glomerular Filtration Rate/drug effects , Nephritis, Interstitial/chemically induced , Peptide Fragments/administration & dosage , Peptide Fragments/toxicity , Acute Disease , Angiogenesis Inhibitors/pharmacokinetics , Animals , Apolipoproteins A/pharmacokinetics , Area Under Curve , Blood Urea Nitrogen , Dose-Response Relationship, Drug , Glomerular Filtration Barrier , Infusions, Intravenous , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Male , Nephritis, Interstitial/pathology , Nephritis, Interstitial/physiopathology , Peptide Fragments/pharmacokinetics , Rats , Rats, Sprague-Dawley , Recovery of Function , Toxicity Tests , Weight Gain/drug effects
13.
Arterioscler Thromb Vasc Biol ; 30(12): 2504-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20966404

ABSTRACT

OBJECTIVE: Apolipoprotein A-V (apoA-V), a minor protein associated with lipoproteins, has a major effect on triacylglycerol (TG) metabolism. We investigated whether apoA-V complexed with phospholipid in the form of a reconstituted high-density lipoprotein (rHDL) has potential utility as a therapeutic agent for treatment of hypertriglyceridemia (HTG) when delivered intravenously. METHODS AND RESULTS: Intravenous injection studies were performed in genetically engineered mouse models of severe HTG, including apoav-/- and gpihbp1-/- mice. Administration of apoA-V rHDL to hypertriglyceridemic apoav-/- mice resulted in a 60% reduction in plasma TG concentration after 4 hours. This decline can be attributed to enhanced catabolism/clearance of very-low-density lipoprotein (VLDL), where VLDL TG and cholesterol were reduced ≈60%. ApoA-V that associated with VLDL after injection was also rapidly cleared. Site-specific mutations in the heparin-binding region of apoA-V (amino acids 186 to 227) attenuated apoA-V rHDL TG-lowering activity by 50%, suggesting that this sequence element is required for optimal TG-lowering activity in vivo. Unlike apoav-/- mice, injection of apoA-V rHDL into gpihbp1-/- mice had no effect on plasma TG levels, and apoA-V remained associated with plasma VLDL. CONCLUSIONS: Intravenously injected apoA-V rHDL significantly lowers plasma TG in an apoA-V deficient mouse model. Its intravenous administration may have therapeutic benefit in human subjects with severe HTG, especially in cases involving apoA-V variants associated with HTG.


Subject(s)
Apolipoproteins A/administration & dosage , Apolipoproteins/deficiency , Hypertriglyceridemia/drug therapy , Hypolipidemic Agents/administration & dosage , Lipoproteins, HDL/administration & dosage , Receptors, Lipoprotein/metabolism , Animals , Apolipoprotein A-V , Apolipoproteins/genetics , Apolipoproteins A/blood , Binding Sites , Biomarkers/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Heparin/administration & dosage , Heparin/metabolism , Humans , Hypertriglyceridemia/genetics , Hypertriglyceridemia/metabolism , Hypolipidemic Agents/blood , Injections, Intravenous , Lipoproteins, HDL/blood , Lipoproteins, VLDL/blood , Male , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Mutation , Receptors, Lipoprotein/genetics , Recombinant Proteins/administration & dosage , Time Factors , Triglycerides/blood
14.
Am J Physiol Regul Integr Comp Physiol ; 296(1): R43-50, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19020287

ABSTRACT

CCK and apolipoprotein AIV (apo AIV) are gastrointestinal satiety signals whose synthesis and secretion by the gut are stimulated by fat absorption. Intraperitoneally administered CCK-8 is more potent in suppressing food intake than a similar dose administered intravenously, but the reason for this disparity is unclear. In contrast, both intravenous and intraperitoneally administered apo AIV are equally as potent in inhibiting food intake. When we compared the lymphatic concentration of CCK-8 and apo AIV, we found that neither intraperitoneally nor intravenously administered CCK-8 or apo AIV altered lymphatic flow rate. Interestingly, intraperitoneal administration of CCK-8 produced a significantly higher lymphatic concentration at 15 min than did intravenous administration. Intraperitoneal injection of apo AIV also yielded a higher lymphatic concentration at 30 min than did intravenous administration. Intraperitoneal administration of CCK-8 and apo AIV also resulted in a much longer period of elevated CCK-8 and apo AIV peptide concentration in lymph than intravenous administration. Furthermore, enzymatic activity of dipeptidyl peptidase IV (DPPIV) and aminopeptidase was higher in plasma than in lymph during fasting, and so, satiation peptides, such as CCK-8 and apo AIV in the lymph, are protected from degradation by the significantly lower DPPIV and aminopeptidase activity levels in lymph than in plasma. Therefore, the higher potency of intraperitoneally administered CCK-8 compared with intravenously administered CCK-8 in inhibiting food intake may be explained by both its higher concentration in lymph and the prolonged duration of its presence in the lamina propria.


Subject(s)
Apolipoproteins A/administration & dosage , Appetite Depressants/administration & dosage , Cholecystokinin/administration & dosage , Eating/drug effects , Intestines/drug effects , Lymph/drug effects , Peptide Fragments/administration & dosage , Aminopeptidases/blood , Animals , Apolipoproteins A/metabolism , Appetite Depressants/metabolism , Cholecystokinin/metabolism , Dipeptidyl Peptidase 4/blood , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Injections, Intravenous , Intestinal Mucosa/metabolism , Lymph/enzymology , Male , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Time Factors
15.
Am J Physiol Gastrointest Liver Physiol ; 286(6): G885-90, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15132947

ABSTRACT

The focus of this article is to review evidence that apolipoprotein A-IV (apo A-IV) acts as a satiety factor. Additionally, information regarding the general involvement of apo A-IV in the regulation of food intake and body weight is stated. Apo A-IV is a glycoprotein synthesized by the human intestine. In rodents, both the small intestine and liver secrete apo A-IV, but the small intestine is the major organ responsible for circulating apo A-IV. There is now solid evidence that the hypothalamus, especially the arcuate nucleus, is another active site of apo A-IV expression. Intestinal apo A-IV synthesis is markedly stimulated by fat absorption and does not appear to be mediated by the uptake or reesterification of fatty acids to form triglycerides. Rather, the local formation of chylomicrons acts as a signal for the induction of intestinal apo A-IV synthesis. Intestinal apo A-IV synthesis is also enhanced by a factor from the ileum, probably peptide tyrosine-tyrosine (PYY). The inhibition of food intake by apo A-IV is mediated centrally. The stimulation of intestinal synthesis and secretion of apo A-IV by lipid absorption are rapid; thus apo A-IV likely plays a role in the short-term regulation of food intake. Other evidence suggests that apo A-IV may also be involved in the long-term regulation of food intake and body weight, as it is regulated by both leptin and insulin. Chronic ingestion of a high-fat diet blunts the intestinal as well as the hypothalamic apo A-IV response to lipid feeding. It also suppresses apo A-IV gene expression in the hypothalamus. Whereas it is tempting to speculate that apo A-IV may play a role in diet-induced obesity, we believe the confirmation of such a proposal awaits further experimental evidence.


Subject(s)
Apolipoproteins A/physiology , Gastrointestinal Tract/physiology , Satiety Response/physiology , Animals , Apolipoproteins A/administration & dosage , Body Weight/physiology , Eating/drug effects , Humans , Infusions, Intravenous , Injections, Intraventricular , Peptide YY/physiology
16.
Annu Rev Nutr ; 21: 231-54, 2001.
Article in English | MEDLINE | ID: mdl-11375436

ABSTRACT

Apolipoprotein A-IV (apo A-IV) is a glycoprotein synthesized by the human intestine. In rodents, both the small intestine and liver secrete apo A-IV, but the small intestine is the major organ responsible for the circulating apo A-IV. Intestinal apo A-IV synthesis is markedly stimulated by fat absorption and appears not to be mediated by the uptake or reesterification of fatty acids to form triglycerides. Rather, the formation of chylomicrons acts as a signal for the induction of intestinal apo A-IV synthesis. Intestinal apo A-IV synthesis is also enhanced by a factor from the ileum, probably peptide tyrosine-tyrosine. The inhibition of food intake by apo A-IV is mediated centrally. The stimulation of intestinal synthesis and the secretion of apo A-IV by lipid absorption are rapid; thus, apo A-IV likely plays a role in the short-term regulation of food intake. Other evidence suggests that apo A-IV may also be involved in the long-term regulation of food intake and body weight. Chronic ingestion of a high-fat diet blunts the intestinal apo A-IV response to lipid feeding and may explain why the chronic ingestion of a high-fat diet predisposes both animals and humans to obesity.


Subject(s)
Apolipoproteins A/physiology , Appetite Regulation/physiology , Eating/physiology , Animals , Apolipoproteins A/administration & dosage , Apolipoproteins A/blood , Apolipoproteins A/metabolism , Dietary Fats/pharmacokinetics , Eating/drug effects , Humans , Intestinal Absorption , Intestines/physiology , Peptide YY/physiology , Satiation/physiology
17.
Int J Pharm ; 214(1-2): 87-91, 2001 Feb 19.
Article in English | MEDLINE | ID: mdl-11282243

ABSTRACT

The in vivo organ distribution of particulate drug carriers is decisively influenced by the interaction with plasma proteins after i.v. administration. Serum protein adsorption on lipid drug conjugate nanoparticles, a new carrier system for i.v. application, was investigated by 2-dimensional electrophoresis (2-DE). The particles were surface-modified to target them to the brain. To assess the protein adsorption pattern after i.v. injection in mice prior to in vivo studies, the particles were incubated in mouse serum. Incubation in human serum was carried out in parallel to investigate similarities or differences in the protein patterns obtained from men and mice. Distinct differences were found. Particles incubated in human serum showed preferential adsorption of apolipoproteins A-I, A-IV and E. Previously, preferential adsorption of ApoE was reported as one important factor for targeting of Tween(R)80 modified polybutylcyanoacrylate nanoparticles to the brain. Preferential adsorption of ApoA-I and A-IV took place after incubation in mouse serum, adsorption of ApoE could not be clearly confirmed. In vivo localization of the LDC nanoparticles at the blood-brain barrier and diffusion of the marker Nile Red into the brain could be shown by confocal laser-scanning microscopy. Differences of the obtained adsorption patterns are discussed with regard to their relevance for correlations of in vitro and in vivo data obtained from different species.


Subject(s)
Blood Proteins/metabolism , Brain/metabolism , Drug Delivery Systems , Adsorption , Animals , Apolipoprotein A-I/administration & dosage , Apolipoproteins A/administration & dosage , Apolipoproteins E/administration & dosage , Blood-Brain Barrier , Humans , Mice , Species Specificity
19.
Circulation ; 100(6): 594-8, 1999 Aug 10.
Article in English | MEDLINE | ID: mdl-10441095

ABSTRACT

BACKGROUND: Apolipoprotein (apo) A-I is the major protein component of HDL, a cholesterol transport particle that protects against atherosclerosis. Apo A-I is believed to promote reverse cholesterol transport, transferring cholesterol from peripheral cells to the liver for subsequent elimination. To test this hypothesis in humans, we measured fecal steroid excretion before and after the intravenous infusion of human proapo A-I (precursor of apo A-I) liposome complexes. METHODS AND RESULTS: Four subjects with heterozygous familial hypercholesterolemia w re studied under standardized conditions. The fecal excretion of bile acids and neutral sterols was determined for 9 days before and 9 days after an intravenous infusion of recombinant human proapo A-I (4 g protein) liposome complexes. Plasma apoA-I and HDL cholesterol levels increased transiently (mean peak concentrations were 64% and 35% above baseline, respectively) during the first 24 hours. Mean lipoprotein lipid and apolipoprotein levels were not different during the 2 collecting periods, however. Serum lathosterol, a precursor of cholesterol whose concentration reflects the rate of cholesterol synthesis in vivo, was also unchanged. The fecal excretion of cholesterol (neutral sterols and bile acids) increased in all subjects (mean increase, +39% and +30%, respectively), corresponding to the removal of approximately 500 mg/d excess cholesterol after infusion. Control infusions with only liposomes in 2 of the patients did not influence lipoprotein pattern or cholesterol excretion. CONCLUSIONS: Infusion of proapoA-I liposomes in humans promotes net cholesterol excretion from the body, implying a stimulation of reverse cholesterol transport. This mechanism may prove useful in the treatment of atherosclerosis.


Subject(s)
Apolipoproteins A/therapeutic use , Arteriosclerosis/drug therapy , Bile Acids and Salts/analysis , Feces/chemistry , Hyperlipoproteinemia Type II/drug therapy , Liposomes/therapeutic use , Protein Precursors/therapeutic use , Sterols/analysis , Adult , Apolipoprotein A-I/blood , Apolipoprotein A-I/genetics , Apolipoproteins A/administration & dosage , Apolipoproteins A/pharmacology , Biological Transport/drug effects , Biomarkers , Cholesterol/blood , Cholesterol, HDL/blood , Coronary Artery Disease/drug therapy , Coronary Artery Disease/metabolism , Drug Evaluation , Heterozygote , Humans , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/metabolism , Infusions, Intravenous , Liposomes/administration & dosage , Liver/metabolism , Male , Middle Aged , Phospholipids/administration & dosage , Protein Precursors/administration & dosage , Protein Precursors/pharmacology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use
20.
Gastroenterology ; 109(5): 1583-8, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7557142

ABSTRACT

BACKGROUND & AIMS: We recently showed that intracisternal injection of apolipoprotein A-IV (apo A-IV), a protein produced by the small intestine in response to fat, inhibits gastric acid secretion. The aim of this study was to investigate the mechanism of acid inhibition by central apo A-IV. METHODS: Gastric acid secretion was determined in pylorus-ligated conscious rats. The effect of intracisternal injection of apo A-IV on gastric acid secretion stimulated by pentagastrin, bethanechol, or intracisternal thyrotropin-releasing hormone (central vagal stimulant) was examined. The effects of vagotomy, indomethacin, and adrenergic blockers on the acid inhibition of apo A-IV were examined to investigate the role of the vagal system, prostaglandin pathways, and adrenergic system. RESULTS: Intracisternal apo A-IV significantly inhibited pentagastrin-, bethanechol-, and thyrotropin-releasing hormone-stimulated gastric acid secretion in a similar fashion. Inhibition of pentagastrin-stimulated acid secretion by apo A-IV still occurred even in vagotomized rats. Yohimbine but not indomethacin or propranolol eliminated apo A-IV--induced inhibition of acid. CONCLUSIONS: Intracisternal apo A-IV inhibits gastric acid secretion through alpha 2-adrenergic receptors. The vagal pathway and the prostaglandin system are not involved in apo A-IV--induced acid inhibition.


Subject(s)
Apolipoproteins A/pharmacology , Gastric Acid/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Apolipoproteins A/administration & dosage , Bethanechol/pharmacology , Brain/drug effects , Depression, Chemical , Indomethacin/pharmacology , Male , Pentagastrin/pharmacology , Rats , Rats, Sprague-Dawley , Thyrotropin-Releasing Hormone/pharmacology , Vagotomy
SELECTION OF CITATIONS
SEARCH DETAIL
...