1
|
Guo Q, Dong Q, Xu W, Zhang H, Zhao X, He W, He Y, Zhao G. Metabolite profiling of camel milk and the fermentation bacteria agent TR1 fermented two types of sour camel milk using LC-MS in relation to their probiotic potentials. Heliyon 2024; 10:e35801. [PMID: 39220917 PMCID: PMC11365327 DOI: 10.1016/j.heliyon.2024.e35801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Camel milk is a nutrient-rich diet and fermentation affects its nutritional value and probiotic function. In this study, sour camel milk and oat jujube sour camel milk were prepared using fermentation bacteria agent TR1, and the metabolites of camel milk, sour camel milk and oat jujube sour camel milk were detected using a non-targeted metabolomics approach using liquid chromatography-mass spectrometry (LC-MS).The results showed that the partial least squares discriminant analysis (PLS-DA) with 100 % accuracy and good predictive power detected 343 components in positive ion mode and 220 components in negative ion mode. The differential metabolites were mainly organic acids, amino acids, esters, vitamins and other substances contained in camel milk.It showed that there were significant differences in the metabolites of camel milk, sour camel milk and oat jujube sour camel milk. Based on the pathway enrichment analysis of the three dairy products in the KEGG database, 12 metabolic pathways mainly involved in the positive ion mode and 20 metabolic pathways mainly involved in the negative ion mode were identified. The main biochemical metabolic pathways and signal transduction pathways of the differential metabolites of the three dairy products were obtained. This study provides theoretical support for improving the nutritional quality and probiotic function of camel milk and fermented camel milk products and provides a basis for the development of relevant processing technologies and products for camel milk and fermented camel milk.
Collapse
Affiliation(s)
- Qingwen Guo
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Biological Manufacturing in Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University, Hohhot, China
| | - Qigeqi Dong
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Biological Manufacturing in Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University, Hohhot, China
| | - Weisheng Xu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Biological Manufacturing in Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiangyu Zhao
- The People's Bank of China Operation Office, China
| | - Wanxiong He
- Inner Mongolia Medical University, Hohhot, China
| | - Yuxing He
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Guofen Zhao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Biological Manufacturing in Inner Mongolia Autonomous Region, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
2
|
Liu F, Chen S, Ming X, Li H, Zeng Z, Lv Y. Sortilin-induced lipid accumulation and atherogenesis are suppressed by HNF1b SUMOylation promoted by flavone of Polygonatum odoratum. J Zhejiang Univ Sci B 2023; 24:998-1013. [PMID: 37961802 PMCID: PMC10646395 DOI: 10.1631/jzus.b2200682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/17/2023] [Indexed: 11/15/2023]
Abstract
This study aims to investigate the impact of hepatocyte nuclear factor 1β (HNF1b) on macrophage sortilin-mediated lipid metabolism and aortic atherosclerosis and explore the role of the flavone of Polygonatum odoratum (PAOA-flavone)-promoted small ubiquitin-related modifier (SUMO) modification in the atheroprotective efficacy of HNF1b. HNF1b was predicted to be a transcriptional regulator of sortilin expression via bioinformatics, dual-luciferase reporter gene assay, and chromatin immunoprecipitation. HNF1b overexpression decreased sortilin expression and cellular lipid contents in THP-1 macrophages, leading to a depression in atherosclerotic plaque formation in low-density lipoprotein (LDL) receptor-deficient (LDLR-/-) mice. Multiple SUMO1-modified sites were identified on the HNF1b protein and co-immunoprecipitation confirmed its SUMO1 modification. The SUMOylation of HNF1b protein enhanced the HNF1b-inhibited effect on sortilin expression and reduced lipid contents in macrophages. PAOA-flavone treatment promoted SUMO-activating enzyme subunit 1 (SAE1) expression and SAE1-catalyzed SUMOylation of the HNF1b protein, which prevented sortilin-mediated lipid accumulation in macrophages and the formation of atherosclerotic plaques in apolipoprotein E-deficient (ApoE-/-) mice. Interference with SAE1 abrogated the improvement in lipid metabolism in macrophage cells and atheroprotective efficacy in vivo upon PAOA-flavone administration. In summary, HNF1b transcriptionally suppressed sortilin expression and macrophage lipid accumulation to inhibit aortic lipid deposition and the development of atherosclerosis. This anti-atherosclerotic effect was enhanced by PAOA-flavone-facilitated, SAE1-catalyzed SUMOylation of the HNF1b protein.
Collapse
Affiliation(s)
- Fang Liu
- Guangxi Key Laboratory of Diabetic Systems Medicine & Institute of Basic Medical Sciences, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541199, China
| | - Shirui Chen
- Guangxi Key Laboratory of Diabetic Systems Medicine & Institute of Basic Medical Sciences, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541199, China
| | - Xinyue Ming
- Guangxi Key Laboratory of Diabetic Systems Medicine & Institute of Basic Medical Sciences, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541199, China
| | - Huijuan Li
- Guangxi Key Laboratory of Diabetic Systems Medicine & Institute of Basic Medical Sciences, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541199, China
| | - Zhaoming Zeng
- Hunan Mingshun Pharmaceutical Co., Ltd., Shaodong 422800, China. ,
| | - Yuncheng Lv
- Guangxi Key Laboratory of Diabetic Systems Medicine & Institute of Basic Medical Sciences, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
3
|
Wang Q, Yesitayi G, Liu B, Siti D, Ainiwan M, Aizitiaili A, Ma X. Targeting metabolism in aortic aneurysm and dissection: from basic research to clinical applications. Int J Biol Sci 2023; 19:3869-3891. [PMID: 37564200 PMCID: PMC10411465 DOI: 10.7150/ijbs.85467] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023] Open
Abstract
Aortic aneurysm and dissection (AAD) are a group of insidious and lethal cardiovascular diseases that characterized by seriously threatening the life and health of people, but lack effective nonsurgical interventions. Alterations in metabolites are increasingly recognized as universal features of AAD because metabolic abnormalities have been identified not only in arterial tissue but also in blood and vascular cells from both patients and animal models with this disease. Over the past few decades, studies have further supported this notion by linking AAD to various types of metabolites such as those derived from gut microbiota or involved in TCA cycle or lipid metabolism. Many of these altered metabolites may contribute to the pathogenesis of AAD. This review aims to illustrate the close association between body metabolism and the occurrence and development of AAD, as well as summarize the significance of metabolites correlated with the pathological process of AAD. This provides valuable insight for developing new therapeutic agents for AAD. Therefore, we present a brief overview of metabolism in AAD biology, including signaling pathways involved in these processes and current clinical studies targeting AAD metabolisms. It is necessary to understand the metabolic mechanisms underlying AAD to provides significant knowledge for AAD diagnosis and new therapeutics for treatment.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Gulinazi Yesitayi
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Bingyan Liu
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Dilixiati Siti
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Mierxiati Ainiwan
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Aliya Aizitiaili
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Xiang Ma
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
4
|
Wei XF, Liu YJ, Li SW, Ding L, Han SC, Chen ZX, Lu H, Wang P, Sun YC. Stress response and tolerance mechanisms of NaHCO 3 exposure based on biochemical assays and multi-omics approach in the liver of crucian carp (Carassius auratus). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114633. [PMID: 36889228 DOI: 10.1016/j.ecoenv.2023.114633] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
The development and utilization of saline-alkaline water, an important backup resource, has received widespread attention. However, the underuse of saline-alkaline water, threatened by the single species of saline-alkaline aquaculture, seriously affects the development of the fishery economy. In this work, a 30-day NaHCO3 stress experimental study combined with analyses of untargeted metabolomics, transcriptome, and biochemical approaches was conducted on crucian carp to provide a better understanding of the saline-alkaline stress response mechanism in freshwater fish. This work revealed the relationships among the biochemical parameters, endogenous differentially expressed metabolites (DEMs), and differentially expressed genes (DEGs) in the crucian carp livers. The biochemical analysis showed that NaHCO3 exposure changed the levels of several physiological parameters associated with the liver, including antioxidant enzymes (SOD, CAT, GSH-Px), MDA, AKP, and CPS. According to the metabolomics study, 90 DEMs are involved in various metabolic pathways such as ketone synthesis and degradation metabolism, glycerophospholipid metabolism, arachidonic acid metabolism, and linoleic acid metabolism. In addition, transcriptomics data analysis showed that a total of 301 DEGs were screened between the control group and the high NaHCO3 concentration group, of which 129 up-regulated genes and 172 down-regulated genes. Overall, NaHCO3 exposure could cause lipid metabolism disorders and induce energy metabolism imbalance in the crucian carp liver. Simultaneously, crucian carp might regulate its saline-alkaline resistance mechanism by enhancing the synthesis of glycerophospholipid metabolism, ketone bodies, and degradation metabolism, at the same time increasing the vitality of antioxidant enzymes (SOD, CAT, GSH-Px) and nonspecific immune enzyme (AKP). Herein, all results will provide new insights into the molecular mechanisms underlying the stress responses and tolerance to saline-alkaline exposure in crucian carp.
Collapse
Affiliation(s)
- Xiao-Feng Wei
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
| | - Ying-Jie Liu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shan-Wei Li
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Lu Ding
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shi-Cheng Han
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Zhong-Xiang Chen
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Hang Lu
- College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
| | - Peng Wang
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China.
| | - Yan-Chun Sun
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
5
|
Multi-omics analysis identifies potential mechanisms by which high glucose accelerates macrophage foaming. Mol Cell Biochem 2023; 478:665-678. [PMID: 36029453 DOI: 10.1007/s11010-022-04542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 08/09/2022] [Indexed: 10/15/2022]
Abstract
Atherosclerotic morbidity is significantly higher in the diabetic population. Hyperglycemia, a typical feature of diabetes, has been proven to accelerate foam cell formation. However, the molecular mechanisms behind this process remain unclear. In this study, LPS and IFN-γ were used to convert THP-1-derived macrophages into M1 macrophages, which were then activated with ox-LDL in either high glucose or normal condition. We identified lipids within macrophages by Oil red O staining and total cholesterol detection. The genes involved in lipid absorption, efflux, inflammation, and metabolism were analyzed using qRT-PCR. The mechanisms of high glucose-induced foam cell formation were further investigated through metabolomics and transcriptomics analysis. We discovered that high glucose speed up lipid accumulation in macrophages (both lipid droplets and total cholesterol increased), diminished lipid efflux (ABCG1 down-regulation), and aggravated inflammation (IL1B and TNF up-regulation). Following multi-omics analysis, it was determined that glucose altered the metabolic and transcriptional profiles of macrophages, identifying 392 differently expressed metabolites and 293 differentially expressed genes, respectively. Joint pathway analysis suggested that glucose predominantly disrupted the glycerolipid, glycerophospholipid, and arachidonic acid metabolic pathways in macrophages. High glucose in the glyceride metabolic pathway, for instance, suppressed the transcription of triglyceride hydrolase (LIPG and LPL), causing cells to deposit excess triglycerides into lipid droplets and encouraging foam cell formation. More importantly, high glucose triggered the accumulation of pro-atherosclerotic lipids (7-ketocholesterol, lysophosphatidylcholine, and glycerophosphatidylcholine). In conclusion, this work elucidated mechanisms of glucose-induced foam cell formation via a multi-omics approach.
Collapse
|
6
|
Effect of the Interaction between Viral PB2 and Host SphK1 on H9N2 AIV Replication in Mammals. Viruses 2022; 14:v14071585. [PMID: 35891566 PMCID: PMC9322132 DOI: 10.3390/v14071585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/11/2022] [Accepted: 07/19/2022] [Indexed: 01/25/2023] Open
Abstract
The H9N2 avian influenza virus (AIV) is currently widespread worldwide, posing a severe threat to the poultry industry and public health. Reassortment is an important way for influenza viruses to adapt to a new host. In 2007, the PB2 gene of H9N2 AIV in China was reassorted, and the DK1-like lineage replaced the F/98-like lineage, forming a dominant genotype of G57. This genotype and its reassortants (such as H7N9, H10N8 and H5N6) showed higher mammalian adaptation, and caused increased human infections. However, the adaptive mechanisms of the DK1-like lineage PB2 gene remain unclear. Here, we confirmed that the PB2 lineage of the H9N2 AIV currently prevalent in China still belongs to the DK1-like lineage and, compared with the previously predominant F/98-like lineage, the DK1-like lineage PB2 gene significantly enhances H9N2 AIV to mammalian adaptation. Through transcriptomic analysis and qRT–PCR and western blot experiments, we identified a host factor, sphingosine kinase 1 (SphK1), that is closely related to viral replication. SphK1 inhibits the replication of DK1-like PB2 gene H9N2 AIV, but the ability of SphK1 protein to bind DK1-like PB2 protein is weaker than that of F/98-like PB2 protein, which may contribute to H9N2 AIV containing the DK1-like PB2 gene to escape the inhibitory effect of host factor SphK1 for efficient infection. This study broadens our understanding of the adaptive evolution of H9N2 AIV and highlights the necessity to pay close attention to the AIV that contains the adaptive PB2 protein in animals and humans.
Collapse
|
7
|
Preservation of Smooth Muscle Cell Integrity and Function: A Target for Limiting Abdominal Aortic Aneurysm Expansion? Cells 2022; 11:cells11061043. [PMID: 35326494 PMCID: PMC8947535 DOI: 10.3390/cells11061043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Abdominal aortic aneurysm (AAA) is a silent, progressive disease with significant mortality from rupture. Whilst screening programmes are now able to detect this pathology early in its development, no therapeutic intervention has yet been identified to halt or retard aortic expansion. The inability to obtain aortic tissue from humans at early stages has created a necessity for laboratory models, yet it is essential to create a timeline of events from EARLY to END stage AAA progression. (2) We used a previously validated ex vivo porcine bioreactor model pre-treated with protease enzyme to create “aneurysm” tissue. Mechanical properties, histological changes in the intact vessel wall, and phenotype/function of vascular smooth muscle cells (SMC) cultured from the same vessels were investigated. (3) The principal finding was significant hyperproliferation of SMC from EARLY stage vessels, but without obvious histological or SMC aberrancies. END stage tissue exhibited histological loss of α-smooth muscle actin and elastin; mechanical impairment; and, in SMC, multiple indications of senescence. (4) Aortic SMC may offer a therapeutic target for intervention, although detailed studies incorporating intervening time points between EARLY and END stage are required. Such investigations may reveal mechanisms of SMC dysfunction in AAA development and hence a therapeutic window during which SMC differentiation could be preserved or reinstated.
Collapse
|