Loading

Annals of Vascular Medicine and Research

Differential Liver Gene Expression in Atherosclerosisresistant and –susceptible Japanese Quail Fed a Cholesterol Enriched Diet

Research Article | Open Access | Volume 7 | Issue 3

  • 1. Avian Research Centre, Faculty of Land and Food Systems, Canada
  • 2. Agriculture and Agri-Food Canada Research Centre, Agassiz, Canada
  • 3. The State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, China
  • 4. School of Biological Sciences, Faculty of Science, Hong Kong, SAR, China
+ Show More - Show Less
Corresponding Authors
Kimberly M, Cheng, Avian Research Centre, Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, BC V6T 1Z4, Canada Tel: 1-604-218-2468;
Abstract

Objectives: (1) To use NGS to examine the differential liver gene expression in Japanese quail fed a cholesterol-enriched diet and to identify the gene systems that may be associated with the development of atherosclerosis, and

(2) To see how divergent genetic selection and diet has modified the expression of these gene systems.

Methods:

1. Liver tissue samples were obtained from Atherosclerosis – resistant (RES) and Atherosclerosis- susceptible (SUS) Japanese quail fed either a control or a cholesterol diet.

2. Total RNA was extracted from the tissue samples and processed for the construction of cDNA libraries.

3. Illumina Mi-Seq system was used for sequencing the quail liver transcriptome libraries. The raw reads were quality trimmed, assembled, and enriched for Differential Expression (DE) analysis.

4. Using the common assembly as reference, DE analysis was conducted using DESeq 2 – Bioconductor. DE transcripts were the blasted against the NCBI Nt database. For each comparison, LogFC was used to separate genes into up- or down-regulation. Pathway enrichment analysis was performed on each of the gene lists using g:Profiler and visualized using Cytoscape v3.6.0 using the Enrichment Map plugin v3.1.

Results: When faced with a high dietary cholesterol challenge, RES upregulated pathways that would be needed to maintain cholesterol and cellular homeostasis. SUS upregulated pathways for cholesterol metabolism and bile acid production, but more significantly upregulated pathways for fatty acid metabolism and attenuation of fatty liver conditions. SUS was not able to maintain lipid homeostasis and developed fatty liver and atherosclerosis.

Conclusion: Epigenetics plays an important role in atherosclerosis. Our previous study found RES and SUS hosting different gut microbiota that would differentially affect the processing of dietary cholesterol. We now have evidence that selective breeding has also changed the bird’s ability to maintain lipid and cellular homeostasis in the liver.

Keywords

Differential gene expression; Cholesterol; Atherosclerosis; Quail model

Citation

Cheng KM, Silversides FG, Liu S, Kim JE, Leung FC (2020) Differential Liver Gene Expression in Atherosclerosis- resistant and –susceptible Japanese Quail Fed a Cholesterol Enriched Diet. Ann Vasc Med Res 7(3): 1111.

ABBREVIATIONS

COPD: Chronic Obstructive Pulmonary Disease; RES: Atherosclerosis-Resistant Quail; SUS: AtherosclerosisSusceptible Quail; RES/CON: RES on Control Diet; RES/CHOL: RES on Cholesterol Diet; SUS/CON: SUS on Control Diet; EUS/CHOL: SUS on Cholesterol Diet; NGS: Next Generation Sequencing; DE: Differential Gene Expression; CE: Cholesteryl Ester; ACAT: AcylCoenzyme A (Coa):Cholesterol Acyltransferase; LCAT: Lecithin Cholesterol Acyltransferase; GO Terms: Gene Ontology Terms; ER: Endoplasmic Reticulum; ERAD: ER-Associated Degradation; PM: Plasma Membrane

INTRODUCTION

Chronic obstructive pulmonary disease (COPD) claimed 3.0 million lives in 2016 and has remained the leading causes of death globally in the last 17 years [1]. Patients with COPD are at increased risk of atherosclerosis and other cardiovascular events. Both conditions are systemic disorders with overlapping mechanisms and pathophysiologic processes. Cardiovascular diseases are an important cause of morbidity and mortality in COPD [2]. Although environmental factors such as diet or smoking play an important role in atherosclerosis development, genetic factors represent consequential determinant of atherosclerotic cardiovascular disease risk. Advances in techniques of molecular genetics have revealed that genetic disorders significantly influence susceptibility to atherosclerotic vascular diseases [3]. Although recent genome-wide association studies (GWAS), have confirmed that mediators of inflammation and perturbed lipid metabolism are major players in cardiovascular disease (CVD) development, much of individual disease heritability remains unexplained by the variants identified through GWAS [4]. There are also limitations in using clinical trials and human subjects for genetic studies involving the interaction between genetic and environment.

The atherosclerosis-resistant (RES) and -susceptible (SUS), Japanese quail is a proven animal model for studying atherosclerosis [5-9]. The two strains were developed by divergent selection from a common foundation population. When challenged with a high cholesterol diet, about 80% of the SUS males will develop atherosclerosis whereas only about 4% of the RES males will [10]. One reason why the Japanese quail is a good model to study atherosclerosis is that it can develop “complex” vascular lesions (focal hemorrhage, calcification and fibrosis) that are very similar to lesions in human [10-12]. Previous studies [10,13], have shown that after cholesterol feeding, plasma cholesterol levels remain high for a significantly longer time in the SUS than in the RES males. In addition, SUS males have fatty livers and higher amounts of liver cholesterol than RES males. Li et al. [8] used RT-PCR to examine the differential expression (DE), of 7 candidate genes (HMGCR, FDFT1, SQLE, DHCR7, ABCG5, ABCG8, and APOA1), in the liver of RES and SUS males with and without dietary cholesterol challenge and reported that RES metabolized and excreted cholesterol faster than SUS. However, the number of genes that they have sampled was small and thus could not draw any conclusion on how the gene system works or how the gene system has been affected by selective breeding. The objective of the present study was to conduct NGS analysis of DE in the liver of RES and SUS quail fed a control and a cholesterolenriched diet.

MATERIALS AND METHODS

All experiments were performed in accordance with protocols reviewed and approved by the UBC Animal Care Committee (Certificate # A12-0087). Liver tissue samples were collected from the same RES and SUS individuals whose intestinal contents were analyzed in Liu et al. [9,14]. The rearing and feeding protocol has been described in detail in Liu et al. [14]. Briefly, 80 RES and 80 SUS were fed a semi-synthetic diet [14], prepared according to the National Research Council (NRC) nutrient requirements standards recommended for Japanese quail (http://www.nap. edu/catalog/2114.html) from hatching to 6 weeks of age. At six weeks of age, they were divided into two dietary treatment groups and fed either the semi-synthetic diet (control) or the semi-synthetic diet with additional cholesterol (0.5% w/w) for another 6 weeks [8]. RES on control diet and on cholesterol diet were designated as RES/CON and RES/CHO, respectively. SUS on control diet and on cholesterol diet were designated as SUS/ CON and SUS/CHO, respectively. Individual birds were identified by numbered wing bands. Both RES and SUS fed the same diet were kept in the same pen. Birds on the alternative diets were kept in a neighboring pen. The two side-by-side pens should have similar environment. At 12 weeks of age, four males from each of the four treatment groups with body weight closest to the mean of the population were euthanized by decapitation. Trunk blood was collected into Vacutainer tubes containing lithium heparin and centrifuged at 4 °C for 10 min at 3,000 × g. Plasma was stored at −20 °C until it was later used for lipid analysis [14]. Livers were then quickly removed and a tissue sample was dissected from the lower part of the right lobe and stored in RNAlater reagent (Cat# AM7023, Ambion, Austin, TX, USA) at -200 C until use. The aortic tree (the brachycephalic arteries to their bifurcations and the aorta to the iliac branching) of each bird was dissected out, opened longitudinally and examined under a 10–30X dissecting microscope for a semi-quantitative scoring [12] of the seriousness of the atherosclerotic lesions on the interior wall. A score of 0 (normal) to 4 (presence of severe atherosclerotic lesions) was assigned by two independent scorers who were blind to the genetic and diet status of the bird.

Total RNA extraction

The liver samples were homogenized in 10 - 20 volumes of TRI Reagent™ Solution (Cat#AM9738, Invitrogen, Carlsbad, CA, USA) and incubated for 5 -10 min at room temperature. After centrifugation at 12,000 x g for 15 min at 4o C, the lower part of the sample was transferred to fresh tubes. Chloroform (SigmaAldrich, Cat# 02487, St. Louis, MO, USA) and 20% of TRI Reagent ™ solution were applied to the sample, mixed well and incubated for 15 min at room temperature. The samples we centrifuged at full speed for 15 min at 4o C and the aqueous phase was transferred to fresh tubes. The supernatant was taken and same volume of pre-chilled isopropanol was applied. After incubation at -20 oC, the sample was centrifuged again at 4o C and the pellet saved. After washing with pre-chilled 75% ethanol, the pellet was briefly air-dried and RNA was dissolved in the RNAse free water (QIAgen Cat#129112, Toronto, ON, Canada), for the Illumina MiSeq platform (Illumina, San Diego, CA, USA). Total RNA content of each sample was measured by using NanoDrop 2000c UVVis Spectrophotometer (Thermo Fisher Scientific Inc. Waltham, MA, USA ), and the quality of RNA samples was assessed by 1% agarose gel electrophoresis and Agilent 2100 Bioanalyzer (Agilent technologies, Santa Clara, CA, USA) before proceeding to further downstream analyses.

cDNA Library Preparation

The cDNA libraries were constructed with the TruSeq RNA Sample Preparation Kit v2 (Illumina, San Diego, CA) following the manufacturer’s protocol using 1 μg of total RNA from each liver. The poly-A-containing mRNA molecules were purified using poly-T oligo-attached magnetic beads and fragmented using divalent cations under elevated temperature, then the first strand cDNA was synthesized using reverse transcriptase and random primers, followed by second-strand cDNA synthesis using DNA polymerase I and RNase H. The cDNA fragments then underwent end repair, addition of a single “A” base, and ligation of the adapters. The ligation products were purified and enriched with PCR to create a final cDNA library. The libraries were quantitated with Qubit 2.0 Fluorometer (Life Technologies, Foster City, CA, USA) and validated with Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA).

Sequencing on Mi-Seq and preprocessing of Raw Reads

Illumina Mi-Seq system (Illumina, SanDiego, CA), was used for sequencing the quail liver transcriptome library using Sequencing by Synthesis (SBS) technology. The 16 libraries were run in one go following manufacturer’s instructions using the Mi-Seq Reagent Kit v2 (Illumina, SanDiego, CA), with 2 x 150 PE sequencing. Mi-Seq Control Software 2.2.0-RTA 1.17.28.0— CASAVA-1.8.2 was used to generate paired-end and single-end data in FastQ format [15]. Raw reads were filtered with Q20 quality trimming to remove low quality reads with average quality score <20 and trimming of low quality bases from the end of reads [14]. Sequence pre-processing software Trimmomatic v0.30 [16], was used to obtain clean paired-end and single-end Mi-Seq data in a FastQ format which was also subjected to quality control using FastQC. The high quality, filtered reads were used for downstream analyses. Velvet Assembler [17], was used for de novo assembly using chicken and turkey genome as reference.

Differential Expression (DE) analysis

In order to perform transcript abundance and DE analysis, sequence assemblies from different samples were merged to serve as the common reference for groups being compared. Transcript abundance and differential expression was determined with RSEM / EBseq software tools (http://deweylab.biostat.wisc.edu/ rsem/README.html). Using the common assembly as reference, DE analysis was done using DESeq 2 – Bioconductor [18]. Genes were considered differentially expressed if PPDE was greater than or equal to 0.95. DE transcripts were the blasted against the NCBI Nt database (https://blast.ncbi.nlm.nih.gov/Blast. cgi?PAGE_TYPE=BlastSearch). Transcripts with a blastn coverage in the first quantile were removed, as these may be spurious hits. In order to perform pathway enrichment analysis, transcripts that did not have a corresponding Gallus gallus (chicken) gene name were removed. For each comparison, LogFC was used to separate genes into up- or down-regulation. Pathway enrichment analysis was performed on each of the gene lists using g:Profiler (PMID:27098042), version r1741_e90_eg37 against the Gallus gallus database with the query ordered by descending logFC for upregulated genes, and ascending logFC for downregulated genes. Significance threshold was set to ‘Benjamini-Hochberg FDR’ [19]. Results of the pathway enrichment was visualized using Cytoscape v3.6.0 using the Enrichment Map plugin v3.1 (PMID: 21085593).

RESULTS AND DISCUSSION

Atherosclerotic lesions on the intimal surface of the aortae

All SUS and RES fed the control diet scored 0. All four SUS on cholesterol diet scored 4, while two RES on the same diet scored 0 and two scored 1 [14].

Plasma lipid parameters

There was a significant diet X genotype interaction in plasma Total Cholesterol (TC) (P<0.006; Table 1),

Table 1: Significant (P<0.006) Diet X Genotype interaction in plasma total cholesterol level (mmol/L) [14].

                                N=16

Genotype*

Diet

RES

SUS

Control

4.75 ± 0.39a

5.29 ± 0.46a

Cholesterol

14.10 ± 1.07a

36.65 ± 6.57b

* RES = Atherosclerosis-resistant quail; SUS = Atherosclerosis-susceptible quail

LDL (P<0.004; Table 2)

Table 2: Significant (P<0.004) Diet X Genotype interaction in plasma LDL level (mmol/L) [14].

N = 16

Genotype*

 Diet

RES

SUS

Control

1.04 ± 0.06a

1.29 ± 0.10a

Cholesterol

8.07 ± 2.09a

32.03 ± 6.42b

* RES = Atherosclerosis-resistant quail; SUS = Atherosclerosis-susceptible quail

levels, and LDL/HDL ratio (P<0.004 Table 3).

Table 3: Significant (P<0.004) Diet X Genotype interaction in plasma LDL/HDL ratio [14].

N = 16

Genotype*

 Diet

RES

SUS

Control

0.34 ± 0.02a

0.38 ± 0.05a

Cholesterol

2.45 ± 0.87a

8.52 ± 1.48b

* RES = Atherosclerosis-resistant quail; SUS = Atheros

SUS fed the cholesterol-enriched diet was significantly higher in these parameters than the other three treatment groups [14].

Differential Expression Analysis

Comparison between RES/CON and RES/CHO: Differentially expressed liver genes when comparing RES/CON and RES/CHO grouped by enriched pathways (GO terms), are listed in Supplementary Table S1. There were 63 genes more expressed and 18 genes less expressed by RES/CHO than RES/ CON. As a result, there were 31 upregulated pathways and 5 downregulated pathways in RES/CHO when compared with RES/CON.

One of the most significantly upregulated pathways was Catalytic Activity (see lower left corner of Figure 1),

Figure 1 Upregulation/downregulation of enriched pathways (GO terms) when RES was challenged with a high cholesterol diet.

Figure 1: Upregulation/downregulation of enriched pathways (GO terms) when RES was challenged with a high cholesterol diet.

which mediates steps in reverse transportation of cholesterol. Cholesterol is needed for cell growth and viability (Pathway Cholesterol biosynthetic process). The prevailing view is that mitochondrial NAD(H) pools tend to be oxidized and important for energy homeostasis, whereas cytosolic NADP(H) pools tend to be highly reduced for reductive biosynthesis (Pathway Oxidoreductase activity, acting on the CH-OH group of donors, NAD or NADP as acceptor). During fasting, acetyl-CoA is used as a precursor for the formation of ketone bodies within mitochondria using NAD+ , but following feeding, it is used as a precursor for cholesterol and lipid synthesis using NADPH as a co-factor [20]. Excess cellular cholesterol is stored as cholesteryl esters (CE). The conversion of cholesterol to CE is catalyzed by the enzyme acyl-coenzyme A (CoA): cholesterol acyltransferase (ACAT). However, chronic accumulation of CE in macrophages causes these cells to appear foamy and is a hallmark of early stages in atherosclerosis. Heterocyclic amides potently inhibited liver ACAT (Pathway heterocycle biosynthetic process) [21, 22]. In addition, lecithin cholesterol acyltransferase (LCAT) catalyzes the esterification of HDL cholesterol causing it to relocate from the surface of the lipoprotein to the core of the particle. The surface of HDL is therefore available to accept more free cholesterol for transportation. HDL was found to also transport endogenous miRNAs, and deliver them to recipient cells [23]. However, HDLcarried miRNAs contribute to altering HDL functions and HDL effect on endothelial cells and macrophages. Down regulation of RNA transport may help maintain the normal properties of HDL cholesterol [24]. In the liver, the scavenger receptor class B1 (SRB1) modulates the selective uptake of HDL CE by hepatocytes. HDL complexes with SR-B1 is endocytosed (Pathway Retrograde transport, endosome to Golgi). CE is hydrolyzed by cholesterol ester esterase (Pathway Hydrolase activity, acting on ester bonds), and secreted as biliary cholesterol or utilized to produce steroid hormones [25]. However, SR-B1 also binds modified LDL and maleylated BSA (M-BSA), and contribute to the development of foam cells [26]. Downregulation of Pathway Macromolecular complex binding may be necessary in cholesterol-filled cells.

Three major apolipoproteins are synthesized in the liver (Pathway Macromolecular complex assembly): apoB, apoA-I, and apoE. ApoB is a large apolipoprotein involved in the transport of triglyceride and cholesterol from the liver to the peripheral tissues [27]. Cholesterol transport into mitochondria is the ratedetermining and hormone-sensitive step in steroid biosynthesis. Mitochondrial translocator protein and the steroidogenic acute regulatory protein were shown to be critical for this process. These two proteins functionally interact to facilitate cholesterol transport and may be part of a larger multimeric mitochondrial complex of proteins assembled (Pathway Macromolecular complex subunit organization) to facilitate the hormone- induced cholesterol transfer into mitochondria [28]. Cholesterol-loaded livers are highly susceptible to TNF-a and Fas-mediated apoptosis by a mechanism that involves mitochondrial oxidative stress [29]. Upregulating DNA-dependent ATPase activity may counter oxidative damages [30]. Na (+)-K (+)-ATPase is involved with the maintenance of ion gradients across the plasma membrane (PM) [31]. Dietary cholesterol can induce genes involved in acute inflammation, including three genes of the serum amyloid A family, three major histocompatibility class II antigen genes, and various cytokine-related genes. Atherosclerosis-resistant C57BL/6ByJ mouse strain was largely resistant to dietary induction of these inflammatory and fibrotic response genes, indicating a negative regulation of gene-specific transcription (Pathway Negative regulation of nucleic acid-templated transcription) by the liver [32].

When challenged with high dietary cholesterol, RES upregulated several pathways related to Lymphocyte mediated immunity (See lower right hand corner of Fig 1). Lymphocytes can be classified into different subtypes: CD4+ TH1 cells; CD4+ Tregs; CD8+ T cells; B cells; NKT cells; NK cells. Microenvironmental cues in the form of cytokines and co-stimulatory triggers guide T cells down different functional routes. Studies have found that subtypes like Tregs and B cells are athero-protective, but other subtypes such as CD4+ T cells, CD8+ T cells, and NKT cells have been shown to accelerate atherosclerosis. Preventing lymphocytes to differentiate (Pathway Negative regulation of lymphocyte differentiation), into and minimizing the proliferation (downregulated Pathway Lymphocyte proliferation) of these subtypes may play a crucial role in slowing atherosclerosis development [33].

In summary, when faced with a high dietary cholesterol challenge, RES upregulated pathways that would maximize lipid metabolism, cholesterol transport and bile acids production to excrete excess cholesterol, upregulate pathways to optimize lymphocyte functions, minimize oxidative stress and damage, and down regulate pathways that would promote foam cell formation.

Comparison between SUS/CON and SUS/CHO: Differentially expressed liver genes when comparing SUS/ CON and SUS/CHO grouped by enriched pathways (GO terms) are listed in Supplementary Table S2. There were 103 more expressed genes and no less expressed genes by SUS/CHO than SUS/CON. As a result, there were 28 upregulated pathways in SUS/CHO when compared with SUS/CON. Surprisingly, only 2 genes, FDFT1 and MLLT4, were similarly more expressed in RES challenged with dietary cholesterol.

One of the most significantly upregulated pathways was cellular catabolic process (Figure 2).

Figure 2 Upregulation/downregulation of enriched Pathways (GO terms) when SUS was challenged with a high cholesterol diet

Figure 2: Upregulation/downregulation of enriched Pathways (GO terms) when SUS was challenged with a high cholesterol diet

Triglycerides from de novo lipogenesis (Pathway Small molecule binding) [34], and from internalized remnant lipoproteins and chylomicrons are packaged in VLDL (Pathway ATP binding) and delivered to peripheral tissues such as adipose and muscle for storage and energy utilization. The liver can also package significant quantities of triglycerides in the form of cytosolic lipid droplets that can be harvested to supplement energy reserves during times of fasting or nutrient deprivation [35]. However, when exposed to a diet rich in lipids, hepatocytes will be overloaded with these fat-storage organelle and develop non-alcoholic fatty liver disease that may result in liver fibrosis.

Cholesterol is oxidized in the liver into a variety of bile acids (Pathway cholesterol metabolic process). High levels of STARD4 (Pathway Cholesterol Binding), increases the synthesis of bile acids and CE in liver hepatocytes. STARD5 (Pathway Cholesterol Binding), binds both cholesterol and 25-hydroxycholesterol and appears to function to redistribute cholesterol to the endoplasmic reticulum (ER) and/or the PM [36]. The PDZ1 domain of the four PDZ domain-containing protein PDZK1 has been reported to bind the C terminus of the HDL SR-BI (Pathway Protein C-terminus binding), and to control hepatic SR-BI expression and function [37]. SR-B1 modulates the selective uptake of HDL CE by hepatocytes.

Mitochondria are capable of both the accumulation of Mg2+ and the release of Mg2+ (Pathway Magnesium ion binding). Magnesium is necessary for the activity of LCAT and lipoprotein lipase (LPL), which lower triglyceride levels and elevate the carrying capacity of HDL [38].

Several redox and oxidant signaling pathways involving cholesterol are at play in fatty liver disease development. These include impairment of the mitochondrial (Pathway Regulation of autophagy of mitochondrion), and lysosomal function (Pathway Lysosomal transport), by cholesterol loading of the inner-cell membranes (Pathway Whole Membrane); formation of cholesterol crystals and hepatocyte degradation; and crownlike structures surrounding degrading hepatocytes, activating Kupffer cells, and evoking inflammation. Cholesterol loading in hepatocytes can result in chronic HIF-1α activity because of the decreased oxygen availability and excessive production of nitric oxide and mitochondrial reactive oxygen species (Pathway Response to reactive oxygen species). The failure to respond to ROS is associated with reduced levels of antioxidants, mitochondrial damage, hepatocyte cell death, activation of the immune system and induction of pro-fibrotic mediators [39,40]

While the ER produces the bulk of the structural phospholipids and cholesterol, together with significant levels of triglyceride and CE with non-structural roles, the PM is a scaffold involving in cell-to-cell communication and the initiation of intracellular signals among other functions (Pathway Endomembrane system organization) [41]. Critical to this function is the PM compartmentalization in lipid rafts that control the localization (Pathway Macromolecule localization), and productive interactions of proteins involved in cell signal propagation (Pathway Protein localization to plasma membrane). The appropriate localization of proteins is crucial because it provides the physiological context for their function. Aberrant localization of proteins contributes to the pathogenesis of many human metabolic and cardiovascular diseases. In addition, cells are divided into compartments limited by other membranes whose integrity and homeostasis are finely controlled (Pathway Establishment of vesicle localization), and which determine the identity and function of the different organelles (Pathway Intracellular Organelle) [42]. Lysosome is a key coordinator in the sorting and delivery of lipids to several membrane compartments. Endogenous lipids are also sorted by the lysosome when it fuses with double-membraned autophagosomes. The role of lysosomes is well established in lipid catabolism and in dietary lipid overload, when cellular lipid content or composition induces changes to alter autophagic activity in a tissue-specific manner.

A growing body of evidence indicates a causal link between lysosomal cholesterol accumulation and inflammation leading to cardiovascular disease and liver steatosis [43]. Disruption of a Golgi system interferes with the modification, sorting and delivery of proteins, and with larger cellular processes such as cell migration, stress responses and apoptosis [44]. C-terminalbinding protein 1 (CTBP1), has dehydrogenase activity and is involved in controlling the equilibrium between tubular and stacked structures in the Golgi complex (Pathway Protein C-terminus binding) [45].

Triglycerides represent the major form of storage and transport fatty acids within cells and in the plasma. The liver is the central organ for fatty acid metabolism (Pathway Regulation of triglyceride metabolic process). Fatty acids accrue in liver by hepatocellular uptake from the plasma (Pathway Lipid transporter activity), and by de novo biosynthesis. Fatty acids are eliminated by oxidation within the cell (Pathway carboxylic acid metabolic process), or by secretion into the plasma within triglyceriderich VLDLs (Pathway Lipid transporter activity). Under normal circumstances, the liver stores only small amounts of fatty acids as triglycerides. When exposed to a high dietary lipid, hepatic fatty acid metabolism is altered (Pathway Peptide metabolic process) [46]. Dysregulated lipid transport and metabolism may lead to the accumulation of triglycerides within hepatocytes, resulting in liver steatosis.

Studies have demonstrated that the autonomic nervous system innervating the liver (Pathway Positive regulation of neuron differentiation), plays a crucial role in regulation of hepatic lipid homeostasis, inflammation and fibrosis. Some of the upregulated pathways identified in individuals with Fatty Liver Syndrome have originally been identified from neural tissues. Additionally, there is growing evidence that neurotrophic factors (Pathway Small molecule binding), can modulate all stages of non-alcoholic fatty liver disease [47]. The synthesis of phosphatidylcholine, the major membrane phospholipid, occurs in axons (Pathway Axons), and its synthesis at this location is required for axonal elongation. Cholesterol is delivered from cell bodies to axons by anterograde transport [48].

Liver fibrosis is characterized by the accumulation of collagen-rich extracellular matrix. E3 ubiquitin ligase (Pathway ligase activity), is involved in collagen synthesis in liver fibrosis. Collagen I is a major component of the extracellular matrix essential for supporting and organizing most tissues. Malformed collagen I chains are removed by retrotranslocation into the cytosol, followed by ER-associated degradation (ERAD), a process that reduces the burden of excess unfolded proteins (Pathway Peptide metabolic process) on the ER. E3 ubiquitin ligases catalyze ubiquitination, which can tag specific proteins for degradation [49].

In summary, SUS upregulated pathways in cholesterol metabolism and bile acid production when challenged by high dietary cholesterol, but the more significant upregulated pathways were for fatty acid metabolism and attenuation of fatty liver conditions.

Comparison between RES/CHO and SUS/CHO: Differentially expressed liver genes grouped by enriched pathways (GO terms) when comparing RES/CHO and SUS/CHO are listed in Supplementary Table S3. There were 216 genes in 32 pathways more expressed in RES/CHO than SUS/CHO. There were 94 genes in 20 pathways more expressed by SUS/CHO than RES/CHO.

In comparison with SUS/CHOL, RES/CHOL pathways for maintaining cholesterol homeostasis and cellular homeostasis [50], were significantly more expressed. One of the highly significantly upregulated pathways was Intracellular membranebounded organelle (Figure 3).

Figure 3 Upregulation/downregulation of enriched Pathways (GO terms) when comparing RES and SUS, both on a high cholesterol diet.

Figure 3: Upregulation/downregulation of enriched Pathways (GO terms) when comparing RES and SUS, both on a high cholesterol diet.

Intracellular organelle includes nucleus, mitochondria, plastids, vacuoles, and vesicles (Pathway Cytoplasmic part). The endomembrane system (Pathways Whole Membrane, Protein kinase C binding, Coated membrane), includes the ER (Pathways ubiquitin-dependent ERAD pathway, ‘de novo’ posttranslational protein folding), Golgi apparatus (Pathway Golgi vesicle transport), and lysosomes (Pathways Catalytic activity, Protein glycosylation). Vesicles also allow the exchange of membrane components with a cell’s PM (Pathways Phosphatidylinositol binding, Protein N-linked glycosylation via asparagine). Intracellular trafficking plays a major role in the proper disposition of internalized cholesterol and in the regulation of cholesterol efflux (Pathways Catabolic process) [41,51].

The nucleus contains the majority of the cellular genome and keeps this information physically separated from other cellular functions (Pathway Nucleoside triphosphate metabolic process). The nucleoplasm is separated from the cytoplasm by the nuclear envelope, a double-bilayer membrane contiguous with the ER. Transport through the envelope is controlled by large multiprotein complexes called nuclear pore complexes (NPCs) (Pathway Protein export from nucleus). One of the most common methods by which cells control protein distribution is by modifying the binding affinity of the localization signal for its corresponding receptor. The nuclear localization of many cell cycle–related proteins is controlled by phosphorylation events in or near the Nuclear Localization Sequence (NLS) [52] (Pathway Positive regulation of protein localization to nucleus).

In order to maintain cholesterol homeostasis, RES/CHOL also upregulated the following pathways in comparison with SUS/ CHOL: Regulation of cholesterol metabolic process; Cholesterol binding; Transferase complex; Caveola; Response to inorganic substance. Lipid homeostasis pathways: negative regulation of fatty acid biosynthetic process; carboxylic acid metabolic process; Acetyltransferase activity;Nucleoside diphosphate phosphorylation. Anti-atherosclerosis pathways: Positive regulation of neutrophil migration; Positive regulation of neutrophil chemotaxis; Positive regulation of interferon-beta production; Anion binding.

In comparison with RES/CHOL, the inadequacy of SUS/CHOL to maintain lipid homeostasis became obvious. Among the many pathways that were more expressed by RES/CHOL, there were 6 pathways that SUS has already upregulated when challenged with high dietary cholesterol. Never the less, pathways that were more expressed by SUS/CHOL than RES/CHOL can be grouped into 3 related areas,

1. Maintaining cholesterol homeostasis: RNA metabolic process; SNARE binding; Protein dephosphorylation; Tethering complex; Protein complex; Protein hydroxilation; AMP metabolic process [53].

2. Promoting lypogenesis and Increasing cellular triglycerides level: Chromatin organization; Organelle membrane fusion; Protein localization to organelle; Oxidoreductase activity, acting on the CH-OH group of donors, NAD or NADP as acceptor; NAD binding.

3. Attenuation of steatosis or atherosclerosis: Rab GTPase binding; Intracellular membrane-bounded organelle; Phosphoprotein phosphatase activity; Regulation of cytokine production involved in immune response; Protein transport; in utero embryonic development (This GO term may be misleading. The genes in this pathway are involved with repressing polymerase II transcription and suppressing cell death) [54].

In summary, RES/CHOL was able to maintain lipid and cellular homeostasis to avoid the development of atherosclerosis, whereas SUS/CHOL was unable to maintain lipid homeostasis and resulting in development of Fatty Liver and atherosclerosis [55].

Comparison between RES/CON and SUS/CON: Differentially expressed liver genes when comparing RES/CON and SUS/CON grouped by enriched pathways (GO terms) are listed in Supplementary Table S4. There were 162 genes in 11 pathways that were more expressed in RES/CON than SUS/CON. There were 33 genes in 1 pathway that were more expressed by SUS/CON than RES/CON (Figure 4).

Figure 4 Upregulation/downregulation of enriched Pathways (GO terms) when comparing RES and SUS, both on control diet

Figure 4: Upregulation/downregulation of enriched Pathways (GO terms) when comparing RES and SUS, both on control diet

The SUS and RES were developed by divergent selection from the same foundation population. The selection criterion was atherosclerotic plaque score after the birds were exposed to dietary cholesterol. The DE when the birds were on control diet was therefore a correlated response to the selection. Pathways that were upregulated by RES/CON in comparison with SUS/CON can be grouped into two related areas:

1. Regulation of lipid metabolism – Cytoplasm; Enzyme binding; Golgi membrane.

2. Tumor suppression, disease or toxin resistance – Cellular metabolic process; Oxidoreductase activity, acting on CHNH group of donars;

ER maintenance --Cellular protein catabolic process; ERAD pathway; Macromolecule localization; Proteolysis involve in cellular protein catabolic process.

DNA damage repair -- Ribonucleoprotein complex biogenesis; Modulation of inflammatory response -- Organelle subcompartment; Enzyme binding.

It is well known that wild type Japanese quail has high disease resistance and high tolerance of toxic chemicals [56- 58]. It is more likely that selection for susceptibility to atherosclerosis has suppressed the expression of the genes in these pathways in SUS/ CON rather than the selection for resistance to atherosclerosis has upregulated these pathways in RES/CON.

Only one pathway, Cellular macromolecule localization, was upregulated in SUS/CON compared with RES/CON. An examination of the genes in this pathway revealed that they were mostly associated with Fatty Liver Syndrome, cancer cell adhesion, migration and metastasis, essential hypertension, and response to mycotoxin.

CONCLUSION

Epigenetics plays an important role in atherosclerosis [59,60]. Liu et al. [9,14], examined the gut microbiota of the same birds used in this study and found that RES and SUS fed the same diets and reared in the same environment were hosting different gut microbiome. They have provided evidence that the microbiome in the ileum and ceca of RES contributed significantly towards the resistance to diet induced atherosclerosis. The epigenetics of the intestinal wall probably created intestinal environment that favors different microbes. The intestinal wall also affects the absorption of dietary cholesterol [61], and the selection for resistance to atherosclerosis is likely to have affected, directly or indirectly, the intestinal pathways involving in the absorption of cholesterol [62]. As such, there may be less dietary cholesterol reaching the RES liver than expected and made it easier for RES to maintain lipid and cellular homeostasis in the liver.

ACKNOWLEDGEMENTS

We thank Dr. Darin Bennett (Avian Research Centre), for project management. The feeding trial was carried out at the Agriculture and Agri-Food Canada Research Station at Agassiz. The NGS sequencing was carried out at the School of Biological Science, University of Hong Kong by Dr Manreet Singh. We would like to thank Vincent Huang (Ontario Institute for Cancer Research), for help with the Bioinformatics and the DE analysis. The funding for this research was provided by the BC Ministry of Agriculture (funds administered by the UBC Specialty Birds Research Committee) to KMC, a grant from the China Agriculture Research System (CARS-42-G13), to SL, and by the Strategic Initiative (Biomedical Engineering) Fund to FCL. SL was also supported by a scholarship from the China Scholarship Council.

REFERENCES

World Health Organization. The top 10 causes of death. 2020.

2.  Chindhi S, Thakur S, Sarkar M, Negi PC. Subclinical atherosclerotic vascular disease in chronic obstructive pulmonary disease: Prospective hospital-based case control study. Lung India. 2015; 32: 137-141.  

3.  Kovacic S, Bakran M. Genetic Susceptibility to Atherosclerosis. Stroke Res Treat. . 2012; 2012: 362941.

4.  Merched AJ, Chan L. Nutrigenetics and nutrigenomics of atherosclerosis. Curr Atheroscler Rep. 2013; 15: 328.

5.  Cheng, KM, Aggrey SE, Nichols CR, Garnett ME, Godin DV. Antioxidant enzymes and atherosclerosis in Japanese quail: Heritability and genetic correlation estimates. Can J Cardiol. 1997; 13: 669-676.

6.  Godin DV, Nichols CR, Hoekstra KA, Garnett ME, Cheng KM. Red cell and plasma antioxidant components and atherosclerosis in Japanese quail: A time-course study. Res Comm Mol Path Pharm. 2001; 110: 27- 51.

7.  Hoekstra KA, Godin DV, Kurtu J, Cheng KM. Heme oxygenase and antioxidant status in cultured aortic endothelial cells isolated from atherosclerosis-susceptible and –resistant Japanese quail. Mol Cell Biochem. 2003; 252: 253-262.

8.  Li X, Schulte P, Godin DV, Cheng KM. Differential mRNA expression of seven genes involved in cholesterol metabolism and transport in the liver of atherosclerosis-susceptible and –resistant Japanese quail strains. Genet Sel Evol. 2012; 44: 20.

9.  Liu S, Tun HM, Leung FC, Bennett DC, Zhang HF, Cheng KM. Interaction of genotype and diet on small intestine microbiota of Japanese quail fed a cholesterol enriched diet” Scien Rep. 2018; 8: 2381.

10. Shih JC, Pullman EP, Kao KJ. Genetic selection, general characterization, and histology of atherosclerosis-susceptible and -resistant Japanese quail. Atherosclerosis. 1983. 49: 41-53.

11. Godin DV, Cheng KM, Garnett ME, Nichols CR. Antioxidant status of Japanese quail: comparison of atherosclerosis-susceptible and –resistant strains. Can J Cardiol. 1994; 10: 221-228.

12. Godin DV, Garnett ME, Cheng KM, Nichols CR. Sex-related alterations in antioxidant status and susceptibility to atherosclerosis in Japanese quail. Can J Cardiol. 1995; 11: 945-951.

13. Godin DV, Nichols CR, Hoekstra KA, Garnett ME, Cheng KM. Alterations in aortic antioxidant components in an experimental model of atherosclerosis: a time-course study. Mol Cell Biochem. 2003; 252: 193-203.

14. Liu S, Bennett DC, Tun HM, Kim JE, Leung FC, Zhang HF, et al. The effect of diet and host genotype on ceca microbiota of Japanese quail fed a cholesterol enriched diet. Front Microb. 2015; 6: 1092.

15. Mazumdar BA, Chattopadhyay S. Sequencing, De novo Assembly, Functional Annotation and Analysis of Phyllanthus amarus Leaf Transcriptome Using the Illumina Platform. Front Plant Sci. 2016; 6: 1199.

16. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014; 30: 2114-2120.

17. Zerbino DR, Birney E. Velvet: algorithms for de novo short read assembly using de Bruijn graphs. Genome Res. 2008; 18: 821-829.

18. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15: 550.

19. Thissen D, Steinberg L, Kuang D. Quick and Easy Implementation of the Benjamini Hochberg Procedure for Controlling the False Positive Rate in Multiple Comparisons. J Educat Behav Stat. 2002; 27: 77-83.

20. Goodman RP, Calvo SE, Mootha VK. Spatiotemporal compartmentalization of hepatic NADH and NADPH metabolism. J Biol Chem. 2018; 293: 7508?7516.

21. White AD, Purchase CF 2nd, Picard JA, Anderson MK, Mueller SB, Bocan TM, et al. Heterocyclic amides: inhibitors of acyl-CoA:cholesterol O-acyl transferase with hypocholesterolemic activity in several species and antiatherosclerotic activity in the rabbit. J Med Chem. 1996; 39: 3908-3919.

22. Chang TY, Li BL, Chang CCY, Urano Y. Acyl-coenzyme A: cholesterol acyltransferases. Am J Physio. 2009; 297: E1-E9.

23. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011; 13: 423-433.  

24. Smolle E, Haybaeck J. Non-coding RNAs and lipid metabolism. Int J Mol Sci. 2014; 15: 13494?13513.  

25. Young IS, Loughrey BV. Lipid-Lowering Therapy. Chapter 88 In: Lip GYH, Hall JE, editors. Comprehensive Hypertension. Elsevier Mosby, Maryland Heights, Missouri. 2007; 1087-1099.

26. Actons, Acton SL, Scherer PE, Lodish HF, Krieger M. Expression cloning of SR-BI, a CD36-related class B scavenger receptor. J Biol Chem. 1994; 269: 21003-21009.

27. Dixon JL, Ginsberg HN. Hepatic synthesis of lipoproteins and apolipoproteins. Semin Liver Dis. 1992; 12: 364-372.

28. Papadopoulos V, Liu J, Culty M. Is there a mitochondrial signaling complex facilitating cholesterol import? Mol Cell Endocrin. 2007; 265-266.

29. Marí M, Caballero F, Colell A, Morales A, Caballeria J, Fernandez A, et al. Mitochondrial free cholesterol loading sensitizes to TNF- and Fas-mediated steatohepatitis. Cell Metab. 2006; 4: 185-198.

30. Caro P, Gomez J, Sanchez I, Naudi A, Ayala V, Lopez-Torres M, et al. Forty Percent Methionine Restriction Decreases Mitochondrial Oxygen Radical Production and Leak at Complex I During Forward Electron Flow and Lowers Oxidative Damage to Proteins and Mitochondrial DNA in Rat Kidney and Brain Mitochondria. Rejuv Res. 2009; 12: 421-434.

31. Buck LT, Hochachka PW, Schön A, Gnaiger E. Microcalorimetric measurement of reversible metabolic suppression induced by anoxia in isolated hepatocytes. Am J Physiol. 1993; 265: R1014-R1019.

32. Vergnes L, Phan J, Strauss M, Tafuri S, Reue K. Cholesterol and Cholate Components of an Atherogenic Diet Induce Distinct Stages of Hepatic Inflammatory Gene Expression. J Biol Chem. 2003; 278: 42774-42784.  

33. Hedrick CC. Lymphocytes in Atherosclerosis. Arterioscler Thromb Vasc Biol. 2015; 35: 253-257.

34. Choi SH, Ginsberg HN. Increased very low density lipoprotein (VLDL) secretion, hepatic steatosis, and insulin resistance. Trends Endocrinol Metab. 2011; 22: 353?363.

35. Olzmann JA, Carvalho P. Dynamics and functions of lipid droplets. Nat Rev Mol Cell Biol. 2019; 20: 137-155.  

36. Wang J, Bie J, Ghosh S. Intracellular cholesterol transport proteins enhance hydrolysis of HDL-CEs and facilitate elimination of cholesterol into bile. J Lipid Res. 2016; 57: 1712?1719.

37. Trigatti BL. SR-B1 and PDZK1: partners in HDL regulation. Curr Opin Lipidol. 2017; 28: 201-208.

38. Pilchova I, Klacanova K, Tatarkova Z, Kaplan P, Racay P. The Involvement of Mg2+ in Regulation of Cellular and Mitochondrial Functions. Oxid Med Cell Long. 2017; 6797460.

39. Tirosh O. Hypoxic Signaling and Cholesterol Lipotoxicity in Fatty Liver Disease Progression. Oxid Med Cell Longev. 2018; 2018: 2548154.

40. Prieto I, Monsalve M. ROS homeostasis, a key determinant in liver ischemic-preconditioning. Redox Biol. 2017; 12: 1020-1025.

41. Lippincott-Schwartz J, Phair RD. Lipids and cholesterol as regulators of traffic in the endomembrane system. Annu Rev Biophys. 2010; 39: 559-578.

42. Hung M-C, Link W. Protein localization in disease and therapy. J Cell Sci. 2011; 124: 3381-3392.

43. Casares D, Escribá PV, Rosselló CA. Membrane Lipid Composition: Effect on Membrane and Organelle Structure, Function and Compartmentalization and Therapeutic Avenues. Int J Mol Sci. 2019; 20: pii E2167.

44. Hendrikx T, Walenbergh SMA, Hofker MH, Shiri?Sverdlov R. Lysosomal cholesterol accumulation: driver on the road to inflammation during atherosclerosis and non?alcoholic steatohepatitis. Obesity Rev. 2014; 15: 424-433.

45. Kumar V, Carlson JE, Ohgi KA, Edwards TA, Rose D, Escalante CR, et al. Transcription corepressor CtBP is an NAD(+)-regulated dehydrogenase. Mol Cell. 2002; 10: 857-869.

46. Alves-Bezerra M, Cohen DE. Triglyceride Metabolism in the Liver. Compr Physiol. 2017; 8: 1-8.

47. Amir M, Yu M, He P, Srinivasan S. Hepatic Autonomic Nervous System and Neurotrophic Factors Regulate the Pathogenesis and Progression of Non-alcoholic Fatty Liver Disease. Front Med (Lausanne). 2020; 7: 62.

48. Vance JE, Campenot RB, Vance DE. The synthesis and transport of lipids for axonal growth and nerve regeneration. Biochim Biophys Acta. 2000; 1486: 84-96.

49. Hasegawa D, Fujii R, Yagishita N, Matsumoto N, Aratani S, Izumi T, et al. E3 ubiquitin ligase synoviolin is involved in liver fibrogenesis. PLoS One. 2010; 5: e13590.  

50. Afonso MS, Machado RM, Lavrador MS, Quintao ECR, Moore KJ, Lottenberg AM. Molecular Pathways Underlying Cholesterol Homeostasis. Nutrients 2018; 10: 760.

51. Litvinov DY, Savushkin EV, Dergunov AD. Intracellular and Plasma Membrane Events in Cholesterol Transport and Homeostasis. J Lipids. 2018; 2018: 3965054

52. Subramaniam PS, Larkin J 3rd, Mujtaba MG, Walter MR, Johnson HM. The COOH-terminal nuclear localization sequence of interferon gamma regulates STAT1 alpha nuclear translocation at an intracellular site. J Cell Sci. 2000; 113: 2771-2781.

53. Kim EJ, Kim B-h, Seo HS, Lee YJ, Kim HH, Son H-H, et al. Cholesterol-Induced Non-Alcoholic Fatty Liver Disease and atherosclerosis Aggravated by Systemic Inflammation. PloS1. 2014; 9: e97841.

54. Taipale T, Seppälä I, Raitoharju E, Mononen N, Lyytikäinen L-P, Illig T, et al. Fatty liver is associated with blood pathways of inflammatory response, immune system activation and prothrombotic state in Young Finns Study. Scientific Reports. 2018; 8: 10358.

55. Völzke H, Robinson DM, Kleine V, Deutscher R, Hoffmann W, Lüdemann J, et al. Hepatic steatosis is associated with an increased risk of carotid atherosclerosis. World J Gastroenterol. 2005; 11: 1848-1853.

56. Nain S, Bour A, Chalmers C, Smits JE. Immunotoxicity and disease resistance in Japanese quail (Corturnix coturnix japonica) exposed to malathion. Ecotoxicology 2011; 20: 892-900.

57. Lewis DC, Metallinos-Katzaras E, Grivetti LE. Coturnism: Human Poisoning by European Migratory Quail. J Cultural Geog. 2009; 19877: 51-65.

58. Bean TG, Gross MS, Karouna-Renier NK, Henry PFP, Schultz SL, Hladik ML, et al. Toxicokinetics of Imidacloprid-Coated Wheat Seeds in Japanese Quail (Coturnix japonica) and an Evaluation of Hazard. Environ. Sci. Technol. 2019; 53: 3888-3897.

59. Turunen MP, Aavik E, Ylä-Herttuala S. Epigenetics and atherosclerosis. Biochim Biophys Acta. 2009; 1790: 886-891.

60. Wise IA, Charchar FJ. Epigenetic Modifications in Essential Hypertension. Int J Mol Sci. 2016; 17: 451-464.

61. Kramer W, Girbig F, Corsiero D, Burger K, Fahrenholz F, Jung C, et al. Intestinal cholesterol absorption: identification of different binding proteins for cholesterol and cholesterol absorption inhibitors in the enterocyte brush border membrane. Biochim Biophys Acta. 2003; 1633: 13-26.

62. Beraza N, Trautwein C. The gut?brain?liver axis: A new option to treat obesity and diabetes. Hepatology 2008; 48: 1011-1013.

Cheng KM, Silversides FG, Liu S, Kim JE, Leung FC (2020) Differential Liver Gene Expression in Atherosclerosis- resistant and –susceptible Japanese Quail Fed a Cholesterol Enriched Diet. Ann Vasc Med Res 7(3): 1111.

Received : 16 May 2020
Accepted : 27 May 2020
Published : 28 May 2020
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X