10 research outputs found

    Southern Folkways Journal Review Number 7

    Get PDF
    This collection of articles relating to Bulloch County begins with an report about a project on Statesboro history done by second graders at Trinity Christian School, followed by an account of the Martin Pittman Laboratory School, “Other Bulloch County Tales,” a record of the family of James R. Bird, and memories of the life of Rubye Akins Anderson by Mary Lawrence Anderson. Also included are two accounts on Brooklet, “How ‘Six Jug’ Became Atlanta’s First Automotive Star,” and research on the Dixon family.https://digitalcommons.georgiasouthern.edu/bchs-pubs/1035/thumbnail.jp

    Gene Therapy in a Humanized Mouse Model of Familial Hypercholesterolemia Leads to Marked Regression of Atherosclerosis

    Get PDF
    Familial hypercholesterolemia (FH) is an autosomal codominant disorder caused by mutations in the low-density lipoprotein receptor (LDLR) gene. Homozygous FH patients (hoFH) have severe hypercholesterolemia leading to life threatening atherosclerosis in childhood and adolescence. Mice with germ line interruptions in the Ldlr and Apobec1 genes (Ldlr(-/-)Apobec1(-/-)) simulate metabolic and clinical aspects of hoFH, including atherogenesis on a chow diet.In this study, vectors based on adeno-associated virus 8 (AAV8) were used to deliver the gene for mouse Ldlr (mLDLR) to the livers of Ldlr(-/-)Apobec1(-/-) mice. A single intravenous injection of AAV8.mLDLR was found to significantly reduce plasma cholesterol and non-HDL cholesterol levels in chow-fed animals at doses as low as 3×10(9) genome copies/mouse. Whereas Ldlr(-/-)Apobec1(-/-) mice fed a western-type diet and injected with a control AAV8.null vector experienced a further 65% progression in atherosclerosis over 2 months compared with baseline mice, Ldlr(-/-)Apobec1(-/-) mice treated with AAV8.mLDLR realized an 87% regression of atherosclerotic lesions after 3 months compared to baseline mice. Immunohistochemical analyses revealed a substantial remodeling of atherosclerotic lesions.Collectively, the results presented herein suggest that AAV8-based gene therapy for FH may be feasible and support further development of this approach. The pre-clinical data from these studies will enable for the effective translation of gene therapy into the clinic for treatment of FH

    Chromatin regulator SMARCAL1 modulates cellular lipid metabolism

    No full text
    Abstract Biallelic mutations of the chromatin regulator SMARCAL1 cause Schimke Immunoosseous Dysplasia (SIOD), characterized by severe growth defects and premature mortality. Atherosclerosis and hyperlipidemia are common among SIOD patients, yet their onset and progression are poorly understood. Using an integrative approach involving proteomics, mouse models, and population genetics, we investigated SMARCAL1’s role. We found that SmarcAL1 interacts with angiopoietin-like 3 (Angptl3), a key regulator of lipoprotein metabolism. In vitro and in vivo analyses demonstrate SmarcAL1’s vital role in maintaining cellular lipid homeostasis. The observed translocation of SmarcAL1 to cytoplasmic peroxisomes suggests a potential regulatory role in lipid metabolism through gene expression. SmarcAL1 gene inactivation reduces the expression of key genes in cellular lipid catabolism. Population genetics investigations highlight significant associations between SMARCAL1 genetic variations and body mass index, along with lipid-related traits. This study underscores SMARCAL1’s pivotal role in cellular lipid metabolism, likely contributing to the observed lipid phenotypes in SIOD patients

    Evaluation of the minimum effective dose of AAV.TBG.<i>mLDLR</i> vector in <i>Ldlr-/-Apobec1-/-</i> Mice.

    No full text
    <p>Amounts of (A) Plasma cholesterol and (B) non-HDL cholesterol were evaluated in <i>Ldlr-/-Apobec1-/-</i> mice up to day 35 after treatment with different doses of AAV8.mLDLR (n = 9 animals per dose group). Each point represents mean ± s.d. *P<0.05, **P<0.01, ***P<0.001. (C) Pooled mouse plasma from AAV-injected <i>Ldlr-/-Apobec1-/-</i> (n = 5, per dose group) were analyzed by FPLC fractionation and the cholesterol content of each fraction was determined. (D) Dose response analysis of Day 60 samples examining cholesterol levels as a function of vector dose. (E) Plasma cholesterol and (F) Alanine transaminase were evaluated in <i>Ldlr-/-Apobec1-/-</i> mice up to day 180 days after treatment with 1×10∧11 GC of AAV8.TBG.<i>mLDLR</i> (n = 10) or 1×10∧11 GC of AAV8.TBG.<i>nLacZ</i> (n = 9). Each point represents mean ± s.d.</p

    AAV8.TBG.<i>mLDLR</i> mediated regression of atherosclerotic lesions in high-fat fed <i>Ldlr-/-Apobec1-/-</i>mice.

    No full text
    <p>(A) En face Sudan IV staining. Mouse aortas were pinned and stained with Sudan IV, which stains neutral lipids. Representative aortas from animals treated with 1×10∧11 of AAV8.TBG.<i>nLacZ,</i> 1×10∧11 of AAV8.TBG.<i>mLDLR</i> at day 60 after vector administration (day 120 on high-fat diet), or at baseline (day 60 on high-fat diet) are shown. (B) The percent Sudan IV staining of the total aortic surface in baseline (n = 10), AAV.TBG.<i>nLacZ</i> (n = 9) and AAV8.TBG.<i>mLDLR</i> (n = 10) was determined as described under <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0013424#s2" target="_blank">Materials and Methods</a>. Aortic roots from these mice were stained with oil red o (C) or hematoxylin and eosin (H&E) (D) 10× magnification. Quantification was conducted on oil red o lesions (E) as described in the <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0013424#s2" target="_blank">materials and methods</a>. Each column represents mean ± s.d. *P<0.05, **P<0.01, ***P<0.001, ‡ P<0.001. (F) H&E stained aortic roots at 40× magnification show a thin fibrous cap and expanded necrotic core in lesions of baseline and AAV8.nLacZ treated mice compared to AAV8.mLDLR injected animals.</p

    Evaluation of AAV8.TBG.<i>mLDLR</i> vector in high fat fed <i>Ldlr-/-Apobec1-/-</i> mice.

    No full text
    <p>Amounts of (A) Plasma cholesterol (B) non-HDL cholesterol, and (C) Alanine transaminase were evaluated in <i>Ldlr-/-Apobec1-/-</i> mice up to day 60 after treatment with 1x10∧11 GC of AAV8.TBG.<i>mLDLR</i> (n = 10) or 1×1011 GC of AAV8.TBG.<i>nLacZ</i> (n = 9). Each point represents mean ± s.d. *P<0.05, ‡ P<0.001.</p

    Evaluation of AAV8 encoding mouse <i>Vldlr</i> or mouse <i>Ldlr</i> in Ldlr-/-Apobec1-/- Mice.

    No full text
    <p>(A) Plasma cholesterol levels in <i>Ldlr-/-Apobec1-/-</i> mice after treatment with AAV8.TBG.<i>mVLDLR</i> or AAV8.TBG.<i>mLDLR</i> (n = 5 animals per dose group). Each point represents mean ± s.d. *P<0.05, **P<0.01, ***P<0.001 (B and C) Pooled mouse plasma from AAV-injected <i>Ldlr-/-Apobec1-/-</i> (n = 5) were analyzed by FPLC fractionation and the cholesterol content of each fraction was determined. (B) Lipoprotein profile of animals injected with 1×10∧12 GC of vector 28 days after treatment. (C) Lipoprotein profile of animals injected with 3×10∧11 GC of vector 28 days after treatment. (D) Plasma ALT levels in <i>Ldlr-/-Apobec1-/-</i> mice after treatment with AAV8.TBG.<i>mVLDLR</i> or AAV.TBG.<i>mLDLR</i> (n = 5 animals per dose group). Each point represents mean ± s.d. At all time points and doses examined, no significant differences in ALT were detected between AAV8.TBG.<i>mLDLR</i> and AAV8.TBG.<i>mVLDLR</i>.</p

    Immunohistochemical analysis of mouse atherosclerotic lesions.

    No full text
    <p>Representative aortic root sections immunostained for the foam cell marker CD68 (A), VCAM-1 (B), or Masson trichrome blue stain for collagen content (C). Original magnification, 40×. Note abundant immunostaining for foam cell marker, CD68 (brown), VCAM-1 adhesion molecules (also brown), and presence of collagen αblue) within lesion in baseline and AAV.TBG.<i>nLacZ</i> injected <i>Ldlr-/-Apobec1-/-</i>animals.</p
    corecore