Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Milton Pryor is active.

Publication


Featured researches published by Milton Pryor.


Journal of Pharmacology and Experimental Therapeutics | 2016

Creation of Apolipoprotein C-II (ApoC-II) Mutant Mice and Correction of Their Hypertriglyceridemia with an ApoC-II Mimetic Peptide

Toshihiro Sakurai; Akiko Sakurai; Boris Vaisman; Marcelo Amar; Chengyu Liu; Scott M. Gordon; Steven K. Drake; Milton Pryor; Maureen Sampson; Ling Yang; Lita Freeman; Alan T. Remaley

Apolipoprotein C-II (apoC-II) is a cofactor for lipoprotein lipase, a plasma enzyme that hydrolyzes triglycerides (TGs). ApoC-II deficiency in humans results in hypertriglyceridemia. We used zinc finger nucleases to create Apoc2 mutant mice to investigate the use of C-II-a, a short apoC-II mimetic peptide, as a therapy for apoC-II deficiency. Mutant mice produced a form of apoC-II with an uncleaved signal peptide that preferentially binds high-density lipoproteins (HDLs) due to a 3-amino acid deletion at the signal peptide cleavage site. Homozygous Apoc2 mutant mice had increased plasma TG (757.5 ± 281.2 mg/dl) and low HDL cholesterol (31.4 ± 14.7 mg/dl) compared with wild-type mice (TG, 55.9 ± 13.3 mg/dl; HDL cholesterol, 55.9 ± 14.3 mg/dl). TGs were found in light (density < 1.063 g/ml) lipoproteins in the size range of very-low-density lipoprotein and chylomicron remnants (40–200 nm). Intravenous injection of C-II-a (0.2, 1, and 5 μmol/kg) reduced plasma TG in a dose-dependent manner, with a maximum decrease of 90% occurring 30 minutes after the high dose. Plasma TG did not return to baseline until 48 hours later. Similar results were found with subcutaneous or intramuscular injections. Plasma half-life of C-II-a is 1.33 ± 0.72 hours, indicating that C-II-a only acutely activates lipolysis, and the sustained TG reduction is due to the relatively slow rate of new TG-rich lipoprotein synthesis. In summary, we describe a novel mouse model of apoC-II deficiency and show that an apoC-II mimetic peptide can reverse the hypertriglyceridemia in these mice, and thus could be a potential new therapy for apoC-II deficiency.


BioMed Research International | 2015

Endothelial Expression of Scavenger Receptor Class B, Type I Protects against Development of Atherosclerosis in Mice.

Boris Vaisman; Tatyana G. Vishnyakova; Lita Freeman; Marcelo Amar; Stephen J. Demosky; Chengyu Liu; John A. Stonik; Maureen Sampson; Milton Pryor; Alexander V. Bocharov; Thomas L. Eggerman; Amy P. Patterson; Alan T. Remaley

The role of scavenger receptor class B, type I (SR-BI) in endothelial cells (EC) was examined in several novel transgenic mouse models expressing SR-BI in endothelium of mice with normal C57Bl6/N, apoE-KO, or Scarb1-KO backgrounds. Mice were also created expressing SR-BI exclusively in endothelium and liver. Endothelial expression of the Tie2-Scarb1 transgene had no significant effect on plasma lipoprotein levels in mice on a normal chow diet but on an atherogenic diet, significantly decreased plasma cholesterol levels, increased plasma HDL cholesterol (HDL-C) levels, and protected mice against atherosclerosis. In 8-month-old apoE-KO mice fed a normal chow diet, the Tie2-Scarb1 transgene decreased aortic lesions by 24%. Mice expressing SR-BI only in EC and liver had a 1.5 ± 0.1-fold increase in plasma cholesterol compared to mice synthesizing SR-BI only in liver. This elevation was due mostly to increased HDL-C. In EC culture studies, SR-BI was found to be present in both basolateral and apical membranes but greater cellular uptake of cholesterol from HDL was found in the basolateral compartment. In summary, enhanced expression of SR-BI in EC resulted in a less atherogenic lipoprotein profile and decreased atherosclerosis, suggesting a possible role for endothelial SR-BI in the flux of cholesterol across EC.


PLOS ONE | 2016

Lipoprotein X Causes Renal Disease in LCAT Deficiency

Alice Ossoli; Edward B. Neufeld; Seth Thacker; Boris Vaisman; Milton Pryor; Lita Freeman; Christine A. Brantner; Irina N. Baranova; Nicolás O. Francone; Stephen J. Demosky; Cecilia Vitali; Monica Locatelli; Mauro Abbate; Carlamaria Zoja; Guido Franceschini; Laura Calabresi; Alan T. Remaley

Human familial lecithin:cholesterol acyltransferase (LCAT) deficiency (FLD) is characterized by low HDL, accumulation of an abnormal cholesterol-rich multilamellar particle called lipoprotein-X (LpX) in plasma, and renal disease. The aim of our study was to determine if LpX is nephrotoxic and to gain insight into the pathogenesis of FLD renal disease. We administered a synthetic LpX, nearly identical to endogenous LpX in its physical, chemical and biologic characteristics, to wild-type and Lcat-/- mice. Our in vitro and in vivo studies demonstrated an apoA-I and LCAT-dependent pathway for LpX conversion to HDL-like particles, which likely mediates normal plasma clearance of LpX. Plasma clearance of exogenous LpX was markedly delayed in Lcat-/- mice, which have low HDL, but only minimal amounts of endogenous LpX and do not spontaneously develop renal disease. Chronically administered exogenous LpX deposited in all renal glomerular cellular and matrical compartments of Lcat-/- mice, and induced proteinuria and nephrotoxic gene changes, as well as all of the hallmarks of FLD renal disease as assessed by histological, TEM, and SEM analyses. Extensive in vivo EM studies revealed LpX uptake by macropinocytosis into mouse glomerular endothelial cells, podocytes, and mesangial cells and delivery to lysosomes where it was degraded. Endocytosed LpX appeared to be degraded by both human podocyte and mesangial cell lysosomal PLA2 and induced podocyte secretion of pro-inflammatory IL-6 in vitro and renal Cxl10 expression in Lcat-/- mice. In conclusion, LpX is a nephrotoxic particle that in the absence of Lcat induces all of the histological and functional hallmarks of FLD and hence may serve as a biomarker for monitoring recombinant LCAT therapy. In addition, our studies suggest that LpX-induced loss of endothelial barrier function and release of cytokines by renal glomerular cells likely plays a role in the initiation and progression of FLD nephrosis.


Molecular Nutrition & Food Research | 2016

Long‐chain monounsaturated fatty acid‐rich fish oil attenuates the development of atherosclerosis in mouse models

Zhi-Hong Yang; Masahiro Bando; Toshihiro Sakurai; Ye Chen; Beatrice Emma-Okon; Bree Wilhite; Daiju Fukuda; Boris Vaisman; Milton Pryor; Yoshiyuki Wakabayashi; Maureen Sampson; Zu-Xi Yu; Akiko Sakurai; Abdalrahman Zarzour; Hiroko Miyahara; Jiro Takeo; Hiroshi Sakaue; Masataka Sata; Alan T. Remaley

SCOPE Fish oil-derived long-chain monounsaturated fatty acids (LCMUFA) containing chain lengths longer than 18 were previously shown to improve cardiovascular disease risk factors in mice. However, it is not known if LCMUFA also exerts anti-atherogenic effects. The main objective of the present study was to investigate the effect of LCMUFA on the development of atherosclerosis in mouse models. METHODS AND RESULTS LDLR-KO mice were fed Western diet supplemented with 2% (w/w) of either LCMUFA concentrate, olive oil, or not (control) for 12 wk. LCMUFA, but not olive oil, significantly suppressed the development of atherosclerotic lesions and several plasma inflammatory cytokine levels, although there were no major differences in plasma lipids between the three groups. At higher doses 5% (w/w) LCMUFA supplementation was observed to reduce pro-atherogenic plasma lipoproteins and to also reduce atherosclerosis in ApoE-KO mice fed a Western diet. RNA sequencing and subsequent qPCR analyses revealed that LCMUFA upregulated PPAR signaling pathways in liver. In cell culture studies, apoB-depleted plasma from LDLR-K mice fed LCMUFA showed greater cholesterol efflux from macrophage-like THP-1 cells and ABCA1-overexpressing BHK cells. CONCLUSION Our research showed for the first time that LCMUFA consumption protects against diet-induced atherosclerosis, possibly by upregulating the PPAR signaling pathway.


Molecular Nutrition & Food Research | 2017

Dietary α‐cyclodextrin reduces atherosclerosis and modifies gut flora in apolipoprotein E‐deficient mice

Toshihiro Sakurai; Akiko Sakurai; Ye Chen; Boris Vaisman; Marcelo Amar; Milton Pryor; Seth Thacker; Xue Zhang; Xujing Wang; Yubo Zhang; Jun Zhu; Zhi-Hong Yang; Lita Freeman; Alan T. Remaley

SCOPE α-Cyclodextrin (α-CD), a cyclic polymer of glucose, has been shown to lower plasma cholesterol in animals and humans; however, its effect on atherosclerosis has not been previously described. METHODS AND RESULTS apoE-knockout mice were fed either low-fat diet (LFD; 5.2% fat, w/w), or Western high fat diet (21.2% fat) containing either no additions (WD), 1.5% α-CD (WDA); 1.5% β-CD (WDB); or 1.5% oligofructose-enriched inulin (WDI). Although plasma lipids were similar after 11 weeks on the WD vs. WDA diets, aortic atherosclerotic lesions were 65% less in mice on WDA compared to WD (P < 0.05), and similar to mice fed the LFD. No effect on atherosclerosis was observed for the other WD supplemented diets. By RNA-seq analysis of 16S rRNA, addition of α-CD to the WD resulted in significantly decreased cecal bacterial counts in genera Clostridium and Turicibacterium, and significantly increased Dehalobacteriaceae. At family level, Comamonadaceae significantly increased and Peptostreptococcaceae showed a negative trend. Several of these bacterial count changes correlated negatively with % atherosclerotic lesion and were associated with increased cecum weight and decreased plasma cholesterol levels. CONCLUSION Addition of α-CD to the diet of apoE-knockout mice decreases atherosclerosis and is associated with changes in the gut flora.


Frontiers in Immunology | 2018

Myeloid-specific deletion of peptidylarginine deiminase 4 (PAD4) mitigates atherosclerosis.

Yudong Liu; Carmelo Carmona-Rivera; Erica Moore; Nickie L. Seto; Jason S. Knight; Milton Pryor; Zhi-Hong Yang; Saskia Hemmers; Alan T. Remaley; Kerri A. Mowen; Mariana J. Kaplan

Increasing evidence suggests that neutrophil extracellular traps (NETs) may play a role in promoting atherosclerotic plaque lesions in humans and in murine models. The exact pathways involved in NET-driven atherogenesis remain to be systematically characterized. To assess the extent to which myeloid-specific peptidylarginine deiminase 4 (PAD4) and PAD4-dependent NET formation contribute to atherosclerosis, mice with myeloid-specific deletion of PAD4 were generated and backcrossed to Apoe−/− mice. The kinetics of atherosclerosis development were determined. NETs, but not macrophage extracellular traps, were present in atherosclerotic lesions as early as 3 weeks after initiating high-fat chow. The presence of NETs was associated with the development of atherosclerosis and with inflammatory responses in the aorta. Specific deletion of PAD4 in the myeloid lineage significantly reduced atherosclerosis burden in association with diminished NET formation and reduced inflammatory responses in the aorta. NETs stimulated macrophages to synthesize inflammatory mediators, including IL-1β, CCL2, CXCL1, and CXCL2. Our data support the notion that NETs promote atherosclerosis and that the use of specific PAD4 inhibitors may have therapeutic benefits in this potentially devastating condition.


Journal of Molecular Medicine | 2016

Erratum to: Low ambient oxygen prevents atherosclerosis

Ju Gyeong Kang; Ho Joong Sung; Marcelo Amar; Milton Pryor; Alan T. Remaley; Michele D. Allen; Audrey Noguchi; Danielle A. Springer; Jaeyul Kwon; Jichun Chen; Ji hoon Park; Ping yuan Wang; Paul M. Hwang

1 Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA 2 College of Health Science, Eulji University, Seongnam, South Korea 3 Cardiovascular-Pulmonary Branch, NHLBI, NIH, Bethesda, MD, USA 4 Murine Phenotyping Core, NHLBI, NIH, Bethesda, MD, USA 5 School of Medicine, ChungnamNational University, Daejeon, South Korea 6 Hematology Branch, NHLBI, NIH, Bethesda, MD, South Korea J Mol Med (2016) 94:287–289 DOI 10.1007/s00109-016-1399-y


Journal of Molecular Medicine | 2016

Low ambient oxygen prevents atherosclerosis.

Ju Gyeong Kang; Ho Joong Sung; Marcelo Amar; Milton Pryor; Alan T. Remaley; Michele D. Allen; Audrey Noguchi; Danielle A. Springer; Jaeyul Kwon; Jichun Chen; Ji hoon Park; Ping yuan Wang; Paul M. Hwang


Atherosclerosis | 2017

Dietary supplementation with long-chain monounsaturated fatty acid isomers decreases atherosclerosis and alters lipoprotein proteomes in LDLr−/− mice

Zhi-Hong Yang; Scott M. Gordon; Denis Sviridov; Shuibang Wang; Robert L. Danner; Milton Pryor; Boris Vaisman; Yuka Shichijo; Nobushige Doisaki; Alan T. Remaley


Atherosclerosis Supplements | 2018

LCAT Overexpression Protects Against the Formation of LpX in a Murine Model of Drug Induced Intrahepatic Cholestasis

Marcelo Amar; Lita Freeman; Takafumi Nishida; Maureen Sampson; Milton Pryor; Alan T. Remaley

Collaboration


Dive into the Milton Pryor's collaboration.

Top Co-Authors

Avatar

Alan T. Remaley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marcelo Amar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Boris Vaisman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lita Freeman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Maureen Sampson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhi-Hong Yang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Akiko Sakurai

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Scott M. Gordon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Toshihiro Sakurai

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Audrey Noguchi

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge