Network


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

Hotspot


Dive into the research topics where Brian W. Parks is active.

Publication


Featured researches published by Brian W. Parks.


Cell Metabolism | 2013

Genetic Control of Obesity and Gut Microbiota Composition in Response to High-Fat, High-Sucrose Diet in Mice

Brian W. Parks; Elizabeth Nam; Elin Org; Emrah Kostem; Frode Norheim; Simon T. Hui; Calvin Pan; Mete Civelek; Christoph Rau; Brian J. Bennett; Margarete Mehrabian; Luke K. Ursell; Aiqing He; Lawrence W. Castellani; Bradley A. Zinker; Mark S. Kirby; Thomas A. Drake; Christian A. Drevon; Rob Knight; Peter S. Gargalovic; Todd G. Kirchgessner; Eleazar Eskin; Aldons J. Lusis

Obesity is a highly heritable disease driven by complex interactions between genetic and environmental factors. Human genome-wide association studies (GWAS) have identified a number of loci contributing to obesity; however, a major limitation of these studies is the inability to assess environmental interactions common to obesity. Using a systems genetics approach, we measured obesity traits, global gene expression, and gut microbiota composition in response to a high-fat/high-sucrose (HF/HS) diet of more than 100 inbred strains of mice. Here we show that HF/HS feeding promotes robust, strain-specific changes in obesity that are not accounted for by food intake and provide evidence for a genetically determined set point for obesity. GWAS analysis identified 11 genome-wide significant loci associated with obesity traits, several of which overlap with loci identified in human studies. We also show strong relationships between genotype and gut microbiota plasticity during HF/HS feeding and identify gut microbial phylotypes associated with obesity.


Genome Research | 2015

Genetic and environmental control of host-gut microbiota interactions

Elin Org; Brian W. Parks; Jong Wha J. Joo; Benjamin Emert; William Schwartzman; Eun Yong Kang; Margarete Mehrabian; Calvin Pan; Rob Knight; Robert P. Gunsalus; Thomas A. Drake; Eleazar Eskin; Aldons J. Lusis

Genetics provides a potentially powerful approach to dissect host-gut microbiota interactions. Toward this end, we profiled gut microbiota using 16s rRNA gene sequencing in a panel of 110 diverse inbred strains of mice. This panel has previously been studied for a wide range of metabolic traits and can be used for high-resolution association mapping. Using a SNP-based approach with a linear mixed model, we estimated the heritability of microbiota composition. We conclude that, in a controlled environment, the genetic background accounts for a substantial fraction of abundance of most common microbiota. The mice were previously studied for response to a high-fat, high-sucrose diet, and we hypothesized that the dietary response was determined in part by gut microbiota composition. We tested this using a cross-fostering strategy in which a strain showing a modest response, SWR, was seeded with microbiota from a strain showing a strong response, A×B19. Consistent with a role of microbiota in dietary response, the cross-fostered SWR pups exhibited a significantly increased response in weight gain. To examine specific microbiota contributing to the response, we identified various genera whose abundance correlated with dietary response. Among these, we chose Akkermansia muciniphila, a common anaerobe previously associated with metabolic effects. When administered to strain A×B19 by gavage, the dietary response was significantly blunted for obesity, plasma lipids, and insulin resistance. In an effort to further understand host-microbiota interactions, we mapped loci controlling microbiota composition and prioritized candidate genes. Our publicly available data provide a resource for future studies.


Gut microbes | 2016

Sex differences and hormonal effects on gut microbiota composition in mice

Elin Org; Margarete Mehrabian; Brian W. Parks; Petia Shipkova; Xiaoqin Liu; Thomas A. Drake; Aldons J. Lusis

ABSTRACT We previously reported quantitation of gut microbiota in a panel of 89 different inbred strains of mice, and we now examine the question of sex differences in microbiota composition. When the total population of 689 mice was examined together, several taxa exhibited significant differences in abundance between sexes but a larger number of differences were observed at the single strain level, suggesting that sex differences can be obscured by host genetics and environmental factors. We also examined a subset of mice on chow and high fat diets and observed sex-by-diet interactions. We further investigated the sex differences using gonadectomized and hormone treated mice from 3 different inbred strains. Principal coordinate analysis with unweighted UniFrac distances revealed very clear effects of gonadectomy and hormone replacement on microbiota composition in all 3 strains. Moreover, bile acid analyses showed gender-specific differences as well as effects of gonodectomy, providing one possible mechanism mediating sex differences in microbiota composition.


Cell Metabolism | 2015

Genetic Architecture of Insulin Resistance in the Mouse

Brian W. Parks; Tamer Sallam; Margarete Mehrabian; Nikolas Psychogios; Simon T. Hui; Frode Norheim; Lawrence W. Castellani; Christoph Rau; Calvin Pan; Jennifer Phun; Zhenqi Zhou; Wen-Pin Yang; Isaac M. Neuhaus; Peter S. Gargalovic; Todd G. Kirchgessner; Mark J. Graham; Richard G. Lee; Peter Tontonoz; Robert E. Gerszten; Andrea L. Hevener; Aldons J. Lusis

Insulin resistance (IR) is a complex trait with multiple genetic and environmental components. Confounded by large differences between the sexes, environment, and disease pathology, the genetic basis of IR has been difficult to dissect. Here we examine IR and related traits in a diverse population of more than 100 unique male and female inbred mouse strains after feeding a diet rich in fat and refined carbohydrates. Our results show dramatic variation in IR among strains of mice and widespread differences between sexes that are dependent on genotype. We uncover more than 15 genome-wide significant loci and validate a gene, Agpat5, associated with IR. We also integrate plasma metabolite levels and global gene expression from liver and adipose tissue to identify metabolite quantitative trait loci (mQTL) and expression QTL (eQTL), respectively. Our results provide a resource for analysis of interactions between diet, sex, and genetic background in IR.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2006

Loss of the Lysophosphatidylcholine Effector, G2A, Ameliorates Aortic Atherosclerosis in Low-Density Lipoprotein Receptor Knockout Mice

Brian W. Parks; Aldons J. Lusis; Janusz H. Kabarowski

Objective—Lysophosphatidylcholine is a major product of low-density lipoprotein (LDL) oxidation and secretory phospholipase A2-mediated lipid hydrolysis within atherosclerotic lesions. The G2A receptor mediates chemotaxis of cultured macrophages and T cells to lysophosphatidylcholine, supporting a pro-atherogenic role for this receptor in vivo. We investigated the ability of G2A to modulate atherosclerosis in mice. Methods and Results—We measured atherosclerosis in G2A+/+ and G2A−/− LDL receptor knockout (LDLR−/−) mice. Consistent with a previous study, early lesion size at the aortic sinus was unaffected by G2A deficiency. However, G2A deficiency attenuated lesion progression at this site (42% to 44% reduction in average lesion area) and led to robust suppression of atherosclerosis throughout the aorta after short and extended periods of diet intervention (reduction in aortic lesion coverage: 62% to 73% at 9 weeks, 75% to 84% at 20 weeks). In G2A−/−LDLR−/− mice, intimal macrophage accumulation at lesion-prone sites of the aorta was significantly reduced in the absence of any detectable effect on T cell recruitment. Examination of lipoprotein profiles revealed elevated levels of circulating high-density lipoprotein (HDL) cholesterol in G2A−/−LDLR−/− mice compared with their G2A+/+LDLR−/− counterparts after extended periods of diet intervention (54% increase in mean HDL cholesterol concentration). Conclusion—G2A provides a pro-atherogenic stimulus in vivo consistent with its chemotactic action but to which a pleiotropy of effects, including modulation of lipoprotein metabolism, may also contribute.


Science Translational Medicine | 2016

Skeletal muscle action of estrogen receptor α is critical for the maintenance of mitochondrial function and metabolic homeostasis in females

Vicent Ribas; Brian G. Drew; Zhenqi Zhou; Jennifer Phun; Nareg Y. Kalajian; Teo Soleymani; Pedram Daraei; Kevin Widjaja; Jonathan Wanagat; Thomas Q. de Aguiar Vallim; Amy H. Fluitt; Steven J. Bensinger; Thuc Le; Caius G. Radu; Julian P. Whitelegge; Simon W. Beaven; Peter Tontonoz; Aldons J. Lusis; Brian W. Parks; Laurent Vergnes; Karen Reue; Harpreet Singh; Jean Chrisostome Bopassa; Ligia Toro; Enrico Stefani; Matthew J. Watt; Simon Schenk; Thorbjorn Akerstrom; Meghan Kelly; Bente Klarlund Pedersen

ERα action in skeletal muscle is involved in the preservation of mitochondrial health and insulin sensitivity in female mice and can serve as a defense against metabolic disease in women. Postmenopausal muscle and mitochondrial mayhem Menopause ushers in a host of changes that range from unpleasant to undesirable. One undesirable shift is a loss of protection against insulin resistance, which brings with it a constellation of consequences in the form of chronic disease associated with metabolic dysfunction. Now, Ribas et al. investigate the mechanism underlying the postmenopausal chinks in a woman’s energy homeostasis armor. The estrogen receptor (ER) is known to participate in the preservation of mitochondrial health and insulin sensitivity in mice, but the precise tissue-specific mechanisms remain unclear. Because skeletal muscle is a main tissue responsible for insulin-stimulated glucose disposal, the authors first showed that ERα expression in muscle correlated with metabolic health in human females. They then created a muscle-specific ERα knockout (MERKO) mouse and found that glucose homeostasis was disrupted, fat accumulation increased, and mitochondrial function impaired. These findings imply that ERα action in skeletal muscle helps maintain mitochondrial function and metabolic homeostasis in females. Impaired estrogen receptor α (ERα) action promotes obesity and metabolic dysfunction in humans and mice; however, the mechanisms underlying these phenotypes remain unknown. Considering that skeletal muscle is a primary tissue responsible for glucose disposal and oxidative metabolism, we established that reduced ERα expression in muscle is associated with glucose intolerance and adiposity in women and female mice. To test this relationship, we generated muscle-specific ERα knockout (MERKO) mice. Impaired glucose homeostasis and increased adiposity were paralleled by diminished muscle oxidative metabolism and bioactive lipid accumulation in MERKO mice. Aberrant mitochondrial morphology, overproduction of reactive oxygen species, and impairment in basal and stress-induced mitochondrial fission dynamics, driven by imbalanced protein kinase A–regulator of calcineurin 1–calcineurin signaling through dynamin-related protein 1, tracked with reduced oxidative metabolism in MERKO muscle. Although muscle mitochondrial DNA (mtDNA) abundance was similar between the genotypes, ERα deficiency diminished mtDNA turnover by a balanced reduction in mtDNA replication and degradation. Our findings indicate the retention of dysfunctional mitochondria in MERKO muscle and implicate ERα in the preservation of mitochondrial health and insulin sensitivity as a defense against metabolic disease in women.


Journal of Biological Chemistry | 2014

Open chromatin profiling in mice livers reveals unique chromatin variations induced by high fat diet

Amy Leung; Brian W. Parks; Juan Du; Candi Trac; Ryan Setten; Yin Chen; Kevin Brown; Aldons J. Lusis; Rama Natarajan; Dustin E. Schones

Background: Metabolic diseases result from a combination of multiple genetic and environmental factors. Results: High fat diet leads to chromatin remodeling in mice liver tissue in a strain-dependent manner. Conclusion: Diet can induce changes in the epigenome thereby contributing to metabolic disease. Significance: Environmental factors can contribute to complex disease progression through modifications to chromatin. Metabolic diseases result from multiple genetic and environmental factors. We report here that one manner in which environmental factors can contribute to metabolic disease progression is through modification to chromatin. We demonstrate that high fat diet leads to chromatin remodeling in the livers of C57BL/6J mice, as compared with mice fed a control diet, and that these chromatin changes are associated with changes in gene expression. We further show that the regions of greatest variation in chromatin accessibility are targeted by liver transcription factors, including HNF4α, CCAAT/enhancer-binding protein α (CEBP/α), and FOXA1. Repeating the chromatin and gene expression profiling in another mouse strain, DBA/2J, revealed that the regions of greatest chromatin change are largely strain-specific and that integration of chromatin, gene expression, and genetic data can be used to characterize regulatory regions. Our data indicate dramatic changes in the epigenome due to diet and demonstrate strain-specific dynamics in chromatin remodeling.


Arthritis & Rheumatism | 2011

Autoimmune-mediated reduction of high-density lipoprotein–cholesterol and paraoxonase 1 activity in systemic lupus erythematosus–prone gld mice

Roshni Srivastava; Shaohua Yu; Brian W. Parks; Leland L. Black; Janusz H. Kabarowski

OBJECTIVE To characterize modifications of high-density lipoprotein (HDL) in autoimmune gld mice that may be relevant to premature atherosclerosis in systemic lupus erythematosus, and to assess their relationship to specific aspects of autoimmune disease. METHODS HDL cholesterol (HDL-C), apolipoprotein A-I (Apo A-I), paraoxonase 1 (PON1) activity, hepatic gene expression, and HDL biogenesis were measured in aging female gld and wild-type congenic mice. Autoantibodies, lymphoid organs, and cytokines were analyzed by enzyme-linked immunosorbent assay, flow cytometry, and multiplex assay, respectively. RESULTS Plasma HDL-C, HDL Apo A-I, and HDL-associated PON1 activity were reduced in aging gld mice in association with the development of autoimmunity, independent of changes in hepatic Apo A-I and PON1 expression or HDL biogenesis. Hepatic induction of the acute-phase reactant serum amyloid A1 resulted in its incorporation into HDL in gld mice. Deletion of the lipid-sensitive receptor G2A in gld mice (G2A-/- gld) attenuated reductions in HDL-C and PON1 activity without altering hepatic Apo A-I and PON1 expression, HDL biogenesis, or levels of acute-phase proinflammatory cytokines. Plasma anti-Apo A-I autoantibodies were elevated in aging gld mice commensurate with detectable increases in Apo A-I immune complexes. Autoantibody levels were lower in aging G2A-/- gld mice compared with gld mice, and anti-Apo A-I autoantibody levels were significantly related to HDL-C concentrations (r=-0.645, P<0.00004) and PON1 activity (r=-0.555, P<0.0007) among autoimmune gld and G2A-/- gld mice. CONCLUSION Autoantibodies against Apo A-I contribute to reducing HDL-C and PON1 activity in autoimmune gld mice independently of hepatic HDL biogenesis, suggesting that functional impairment and premature clearance of HDL immune complexes may be principal mechanisms involved.


eLife | 2015

The genetic architecture of NAFLD among inbred strains of mice

Simon T. Hui; Brian W. Parks; Elin Org; Frode Norheim; Nam Che; Calvin Pan; Lawrence W. Castellani; Sarada Charugundla; Darwin L. Dirks; Nikolaos Psychogios; Isaac M. Neuhaus; Robert E. Gerszten; Todd G. Kirchgessner; Peter S. Gargalovic; Aldons J. Lusis

To identify genetic and environmental factors contributing to the pathogenesis of non-alcoholic fatty liver disease, we examined liver steatosis and related clinical and molecular traits in more than 100 unique inbred mouse strains, which were fed a diet rich in fat and carbohydrates. A >30-fold variation in hepatic TG accumulation was observed among the strains. Genome-wide association studies revealed three loci associated with hepatic TG accumulation. Utilizing transcriptomic data from the liver and adipose tissue, we identified several high-confidence candidate genes for hepatic steatosis, including Gde1, a glycerophosphodiester phosphodiesterase not previously implicated in triglyceride metabolism. We confirmed the role of Gde1 by in vivo hepatic over-expression and shRNA knockdown studies. We hypothesize that Gde1 expression increases TG production by contributing to the production of glycerol-3-phosphate. Our multi-level data, including transcript levels, metabolite levels, and gut microbiota composition, provide a framework for understanding genetic and environmental interactions underlying hepatic steatosis. DOI: http://dx.doi.org/10.7554/eLife.05607.001


Radiation Research | 2011

Iron-Ion Radiation Accelerates Atherosclerosis in Apolipoprotein E-Deficient Mice

Tao Yu; Brian W. Parks; Shaohua Yu; Roshni Srivastava; Kiran Gupta; Xing Wu; Saman Khaled; Polly Y. Chang; Janusz H. Kabarowski; Dennis F. Kucik

Abstract Radiation exposure from a number of terrestrial sources is associated with an increased risk for atherosclerosis. Recently, concern over whether exposure to cosmic radiation might pose a similar risk for astronauts has increased. To address this question, we examined the effect of 2 to 5 Gy iron ions (56Fe), a particularly damaging component of cosmic radiation, targeted to specific arterial sites in male apolipoprotein E-deficient (apoE−/−) mice. Radiation accelerated the development of atherosclerosis in irradiated portions of the aorta independent of any systemic effects on plasma lipid profiles or circulating leukocytes. Further, radiation exposure resulted in a more rapid progression of advanced aortic root lesions, characterized by larger necrotic cores associated with greater numbers of apoptotic macrophages and reduced lesional collagen compared to sham-treated mice. Intima media thickening of the carotid arteries was also exacerbated. Exposure to 56Fe ions can therefore accelerate the development of atherosclerotic lesions and promote their progression to an advanced stage characterized by compositional changes indicative of increased thrombogenicity and instability. We conclude that the potential consequences of radiation exposure for astronauts on prolonged deep-space missions are a major concern. Knowledge gained from further studies with animal models should lead to a better understanding of the pathophysiological effects of accelerated ion radiation to better estimate atherogenic risk and develop appropriate countermeasures to mitigate its damaging effects.

Collaboration


Dive into the Brian W. Parks's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Calvin Pan

University of California

View shared research outputs
Top Co-Authors

Avatar

Janusz H. Kabarowski

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Eleazar Eskin

University of California

View shared research outputs
Top Co-Authors

Avatar

Elin Org

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roshni Srivastava

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Shaohua Yu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Simon T. Hui

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge