1
|
Zhou YL, Wu J, Wang HL, Feng WW, Peng F, Zhang RQ, Yan HL, Liu J, Tan YZ, Peng C. Fuzi lizhong pills alter microbial community compositions and metabolite profiles in ulcerative colitis rat with spleen-kidney yang deficiency syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118645. [PMID: 39089661 DOI: 10.1016/j.jep.2024.118645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/06/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is a chronic inflammatory bowel condition that is frequently related with Spleen-Kidney Yang Deficiency Syndrome (SKYD) in Chinese medicine. Fuzi Lizhong Pill (FLZP), a traditional medicine for SKYD, has been utilized in China for generations, although the exact mechanism by which it treats UC is unknown. AIM OF THE STUDY The goal of this study is to further understand FLZP's therapeutic mechanism in SKYD-associated UC. MATERIALS AND METHODS To investigate the impact of FLZP on SKYD-associated UC, we used a comprehensive method that included serum metabolomics and gut microbiota profiling. The chemical composition of FLZP was determined using mass spectrometry. UC rats with SKYD were induced and treated with FLZP. Serum metabolomics and 16S rRNA microbial community analysis were used to evaluate FLZP's effects on endogenous metabolites and gut microbiota, respectively. Correlation analysis investigated the association between metabolites and intestinal flora. A metabolic pathway analysis was undertaken to discover putative FLZP action mechanisms. RESULTS FLZP contains 109 components, including liquiritin (584.8176 μg/g), benzoylaconine (16.3087 μg/g), benzoylhypaconine (31.9583), and hypaconitine (8.1160 μg/g). FLZP predominantly regulated seven metabolites and eight metabolic pathways involved in amino acid and nucleotide metabolism, with an emphasis on energy metabolism and gastrointestinal digestion. FLZP also influenced intestinal flora variety, increasing probiotic abundance while decreasing pathogenic bacteria prevalence. An integrated investigation identified associations between changes in certain gut flora and energy metabolism, specifically the tricarboxylic acid (TCA) cycle. CONCLUSIONS FLZP successfully cures UC in SKYD rats by regulating amino acid and energy metabolism. Its positive effects may include altering microbiota composition and metabolite profiles in UC rats with SKYD. These findings shed light on FLZP's mode of action and its implications for UC management.
Collapse
Affiliation(s)
- Yin-Lin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Pharmacy Department, Zigong Traditional Chinese Medicine Hospital, 643011, China
| | - Jing Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hong-Liang Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wu-Wen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China.
| | - Ruo-Qi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hong-Ling Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Juan Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yu-Zhu Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
2
|
Guo Q, Xie M, Wang QN, Li J, Liu S, Wang X, Yu D, Zou Z, Gao G, Zhang Q, Hao F, Feng J, Yang R, Wang M, Fu H, Bao X, Duan L. Comprehensive Serum Proteomic and Metabolomic Profiles of Pediatric Patients with Moyamoya Disease Reveal Core Pathways. J Inflamm Res 2024; 17:6173-6192. [PMID: 39281778 PMCID: PMC11397188 DOI: 10.2147/jir.s471538] [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: 06/28/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Background Moyamoya disease (MMD) signifies a cerebrovascular disorder with obscure origin and a more rapid and severe progression in children than adults. This investigation aims to uncover age-associated distinctions through proteomic and metabolomic profiling to gain insights into the underlying mechanisms of MMD. Methods Twelve MMD patients-six children and six adults-along with six healthy controls (HC), participated, each providing a 10 mL blood sample. Serum proteomic and metabolomic analyses were conducted using ultra-performance liquid chromatography and high-resolution mass spectrometry, complemented by bioinformatics to identify differential biomolecules and their interactions. Pathway implications were ascertained using GO and KEGG enrichment analysis. Results Notable proteomic and metabolomic discrepancies were observed between pediatric and adult MMD subjects. A total of 235 and 216 proteins varied in adult and pediatric cases compared to HCs, with 73 proteins shared. In addition, 129 and 74 anionic, plus 96 and 104 cationic metabolites, were differentially expressed in the pediatric and adult groups, respectively, with 34 anionic and 28 cationic metabolites in common. Age-specific biomolecules further characterized these distinctions. Enrichment analysis pinpointed immunity and inflammation pathways, with vitamin digestion and absorption highlighted as pivotal in pediatric MMD. Conclusion This study unveils distinct metabolic and proteomic patterns within pediatric and adult MMD patients. The critical role of the vitamin digestion and absorption pathway in the pathogenesis of pediatric MMD offers novel insight into disease mechanisms.
Collapse
Affiliation(s)
- Qingbao Guo
- Medical School of Chinese PLA, Beijing, People's Republic of China
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Manli Xie
- Department of Occupational Diseases, Xi'an Central Hospital, Xi'an, Shanxi, People's Republic of China
| | - Qian-Nan Wang
- Department of Neurosurgery, Eighth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jingjie Li
- Medical School of Chinese PLA, Beijing, People's Republic of China
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Simeng Liu
- Medical School of Chinese PLA, Beijing, People's Republic of China
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiaopeng Wang
- Medical School of Chinese PLA, Beijing, People's Republic of China
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Dan Yu
- Department of Neurosurgery, Fifth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhengxing Zou
- Department of Neurosurgery, Fifth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Gan Gao
- Medical School of Chinese PLA, Beijing, People's Republic of China
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Qian Zhang
- Department of Neurosurgery, Fifth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Fangbin Hao
- Medical School of Chinese PLA, Beijing, People's Republic of China
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jie Feng
- Department of Neurosurgery, Fifth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Rimiao Yang
- Department of Neurosurgery, Fifth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Minjie Wang
- Medical School of Chinese PLA, Beijing, People's Republic of China
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Heguan Fu
- Department of Neurosurgery, Fifth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiangyang Bao
- Department of Neurosurgery, Fifth Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lian Duan
- Department of Neurosurgery, First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
3
|
Laveriano-Santos EP, Luque-Corredera C, Trius-Soler M, Lozano-Castellón J, Dominguez-López I, Castro-Barquero S, Vallverdú-Queralt A, Lamuela-Raventós RM, Pérez M. Enterolignans: from natural origins to cardiometabolic significance, including chemistry, dietary sources, bioavailability, and activity. Crit Rev Food Sci Nutr 2024:1-21. [PMID: 38952149 DOI: 10.1080/10408398.2024.2371939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The enterolignans, enterolactone and enterodiol, the main metabolites produced from plant lignans by the gut microbiota, have enhanced bioavailability and activity compared to their precursors, with beneficial effects on metabolic and cardiovascular health. Although extensively studied, the biosynthesis, cardiometabolic effects, and other therapeutic implications of mammalian lignans are still incompletely understood. The aim of this review is to provide a comprehensive overview of these phytoestrogen metabolites based on up-to-date information reported in studies from a wide range of disciplines. Established and novel synthetic strategies are described, as are the various lignan precursors, their dietary sources, and a proposed metabolic pathway for their conversion to enterolignans. The methodologies used for enterolignan analysis and the available data on pharmacokinetics and bioavailability are summarized and their cardiometabolic bioactivity is explored in detail. The special focus given to research on the health benefits of microbial-derived lignan metabolites underscores the critical role of lignan-rich diets in promoting cardiovascular health.
Collapse
Affiliation(s)
- Emily P Laveriano-Santos
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | | | - Marta Trius-Soler
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Julian Lozano-Castellón
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Inés Dominguez-López
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Sara Castro-Barquero
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, Barcelona, Spain
| | - Anna Vallverdú-Queralt
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Rosa M Lamuela-Raventós
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Maria Pérez
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Zhang Y, Wei S, Zhang H, Jo Y, Kang JS, Ha KT, Joo J, Lee HJ, Ryu D. Gut microbiota-generated metabolites: missing puzzles to hosts' health, diseases, and aging. BMB Rep 2024; 57:207-215. [PMID: 38627947 PMCID: PMC11139682] [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: 10/17/2023] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 05/25/2024] Open
Abstract
The gut microbiota, an intricate community of bacteria residing in the gastrointestinal system, assumes a pivotal role in various physiological processes. Beyond its function in food breakdown and nutrient absorption, gut microbiota exerts a profound influence on immune and metabolic modulation by producing diverse gut microbiota-generated metabolites (GMGMs). These small molecules hold potential to impact host health via multiple pathways, which exhibit remarkable diversity, and have gained increasing attention in recent studies. Here, we elucidate the intricate implications and significant impacts of four specific metabolites, Urolithin A (UA), equol, Trimethylamine N-oxide (TMAO), and imidazole propionate, in shaping human health. Meanwhile, we also look into the advanced research on GMGMs, which demonstrate promising curative effects and hold great potential for further clinical therapies. Notably, the emergence of positive outcomes from clinical trials involving GMGMs, typified by UA, emphasizes their promising prospects in the pursuit of improved health and longevity. Collectively, the multifaceted impacts of GMGMs present intriguing avenues for future research and therapeutic interventions. [BMB Reports 2024; 57(5): 207-215].
Collapse
Affiliation(s)
- Yan Zhang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon 16419, Korea
| | - Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea, Busan 49241, Korea
| | - Hang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, China, Busan 49241, Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea, Busan 49241, Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon 16419, Korea
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea
| | - Jongkil Joo
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Busan 49241, Korea
| | - Hyun Joo Lee
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Busan 49241, Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea, Busan 49241, Korea
| |
Collapse
|
5
|
Li Y, Pan J, Yu JJJ, Wu X, Yang G, Pan X, Sui G, Wang M, Cheng M, Zhu S, Tai H, Xiao H, Xu L, Wu J, Yang Y, Tang J, Gong L, Jia L, Min D. Huayu Qutan Recipe promotes lipophagy and cholesterol efflux through the mTORC1/TFEB/ABCA1-SCARB1 signal axis. J Cell Mol Med 2024; 28:e18257. [PMID: 38526033 PMCID: PMC10962127 DOI: 10.1111/jcmm.18257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/26/2024] Open
Abstract
This study aims to investigate the mechanism of the anti-atherosclerosis effect of Huayu Qutan Recipe (HYQT) on the inhibition of foam cell formation. In vivo, the mice were randomly divided into three groups: CTRL group, MOD group and HYQT group. The HYQT group received HYQT oral administration twice a day (20.54 g/kg/d), and the plaque formation in ApoE-/- mice was observed using haematoxylin-eosin (HE) staining and oil red O (ORO) staining. The co-localization of aortic macrophages and lipid droplets (LDs) was examined using fluorescent labelling of CD11b and BODIPY fluorescence probe. In vitro, RAW 264.7 cells were exposed to 50 μg/mL ox-LDL for 48 h and then treated with HYQT for 24 h. The accumulation of LDs was evaluated using ORO and BODIPY. Cell viability was assessed using the CCK-8 assay. The co-localization of LC3b and BODIPY was detected via immunofluorescence and fluorescence probe. LysoTracker Red and BODIPY 493/503 were used as markers for lysosomes and LDs, respectively. Autophagosome formation were observed via transmission electron microscopy. The levels of LC3A/B II/LC3A/B I, p-mTOR/mTOR, p-4EBP1/4EBP1, p-P70S6K/P70S6K and TFEB protein level were examined via western blotting, while SQSTM1/p62, Beclin1, ABCA1, ABCG1 and SCARB1 were examined via qRT-PCR and western blotting. The nuclear translocation of TFEB was detected using immunofluorescence. The components of HYQT medicated serum were determined using Q-Orbitrap high-resolution MS analysis. Molecular docking was employed to identify the components of HYQT medicated serum responsible for the mTOR signalling pathway. The mechanism of taurine was illustrated. HYQT has a remarkable effect on atherosclerotic plaque formation and blood lipid level in ApoE-/- mice. HYQT decreased the co-localization of CD11b and BODIPY. HYQT (10% medicated serum) reduced the LDs accumulation in RAW 264.7 cells. HYQT and RAPA (rapamycin, a mTOR inhibitor) could promote cholesterol efflux, while chloroquine (CQ, an autophagy inhibitor) weakened the effect of HYQT. Moreover, MHY1485 (a mTOR agonist) also mitigated the effects of HYQT by reduced cholesterol efflux. qRT-PCR and WB results suggested that HYQT improved the expression of the proteins ABCA1, ABCG1 and SCARB1.HYQT regulates ABCA1 and SCARB1 protein depending on the mTORC1/TFEB signalling pathway. However, the activation of ABCG1 does not depend on this pathway. Q-Orbitrap high-resolution MS analysis results demonstrated that seven core compounds have good binding ability to the mTOR protein. Taurine may play an important role in the mechanism regulation. HYQT may reduce cardiovascular risk by promoting cholesterol efflux and degrading macrophage-derived foam cell formation. It has been observed that HYQT and ox-LDL regulate lipophagy through the mTOR/TFEB signalling pathway, rather than the mTOR/4EBP1/P70S6K pathway. Additionally, HYQT is found to regulate cholesterol efflux through the mTORC1/TFEB/ABCA1-SCARB1 signal axis, while taurine plays a significant role in lipophagy.
Collapse
Affiliation(s)
- Yue Li
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio‐Cerebrovascular DiseasesShenyangChina
| | - Jiaxiang Pan
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
- Graduate School of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - J. J. Jiajia Yu
- Postdoctoral Program of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Xize Wu
- Graduate School of Liaoning University of Traditional Chinese MedicineShenyangChina
- Nantong Hospital of Traditional Chinese MedicineNantong Hospital Affiliated to Nanjing University of Chinese MedicineNantongChina
| | - Guanlin Yang
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Xue Pan
- Graduate School of Liaoning University of Traditional Chinese MedicineShenyangChina
- Dazhou Vocational College of Chinese MedicineDazhouChina
| | - Guoyuan Sui
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Mingyang Wang
- College of Animal Science and Veterinary Medicine of Shenyang Agricultural UniversityShenyangChina
| | - Meijia Cheng
- Experimental Center of Traditional Chinese Medicinethe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Shu Zhu
- Department of Paediatric Dentistry, School of StomatologyChina Medical UniversityShenyangChina
| | - He Tai
- School of PharmacyLiaoning University of Traditional Chinese MedicineDalianChina
| | - Honghe Xiao
- School of PharmacyLiaoning University of Traditional Chinese MedicineDalianChina
| | - Lili Xu
- Department of Cardiology, 924 Hospital of Joint Logistic Support Force of PLAGuilinChina
| | - Jin Wu
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Yongju Yang
- Experimental Center of Traditional Chinese Medicinethe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Jing Tang
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| | - Lihong Gong
- Department of Cardiologythe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
- Liaoning Provincial Key Laboratory of TCM Geriatric Cardio‐Cerebrovascular DiseasesShenyangChina
| | - Lianqun Jia
- Innovation Engineering Technology Center of Traditional Chinese MedicineLiaoning University of Traditional Chinese MedicineShenyangChina
| | - Dongyu Min
- Experimental Center of Traditional Chinese Medicinethe Affiliated Hospital of Liaoning University of Traditional Chinese MedicineShenyangChina
| |
Collapse
|
6
|
Matsuo H, Kawakami K, Ohara H, Kaneko T, Mashimo T, Yamada T, Nabika T. Apolipoprotein E-depletion accelerates arterial fat deposition in the spontaneously hypertensive rat. Exp Anim 2023; 72:439-445. [PMID: 37081644 PMCID: PMC10658095 DOI: 10.1538/expanim.23-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/09/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension and atherosclerosis are often found in one patient causing serious cardiovascular events. An animal model simultaneously expressing hypertension and atherosclerosis would be useful to study such a complex risk status. We therefore attempted to introduce a null mutation of the apolipoprotein E (ApoE) gene into the spontaneously hypertensive rat (SHR) using CRISPR/Cas9 to establish a genetic model for atherosclerosis with hypertension. We successfully established SHRApoE(-/-) having a 13-bps deletion in the 5'-end of ApoE gene. Deletion of ApoE protein was confirmed by Western blotting. Blood pressure of SHRApoE(-/-) was comparable to that of SHR. Feeding the rats with high fat high cholesterol diet (HFD) caused a significant increase in LDL cholesterol as well as in triglyceride in SHRApoE(-/-). After 8 weeks of HFD loading, superficial fat deposition was observed both in the aorta and the mesenteric arteries of SHRApoE(-/-) instead of mature atheromatous lesions found in humans. In addition, a null mutation of peroxiredoxin 2 (Prdx2) was introduced into SHRApoE(-/-) to examine the effect of increased oxidative stress on the development of atherosclerosis. SHR with the double depletion of ApoE and Prdx2 did not show mature atheroma either. Further, salt loading did not promote development of atheroma although it accelerated the development of fat deposition. These results indicated that when compared with ApoE-knockout mice, SHRApoE(-/-) was more resistant to atherosclerosis even though they have severe hypertension.
Collapse
Affiliation(s)
- Hiroyuki Matsuo
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
- Department of Functional Pathology, Shimane University Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Kohei Kawakami
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Hiroki Ohara
- Department of Functional Pathology, Shimane University Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Takehito Kaneko
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Tomoji Mashimo
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyou-ku, Kyoto 606-8501, Japan
| | - Takaya Yamada
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Toru Nabika
- Department of Functional Pathology, Shimane University Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| |
Collapse
|
7
|
Deng B, Tao L, Wang Y. Natural products against inflammation and atherosclerosis: Targeting on gut microbiota. Front Microbiol 2022; 13:997056. [PMID: 36532443 PMCID: PMC9751351 DOI: 10.3389/fmicb.2022.997056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/25/2022] [Indexed: 09/29/2023] Open
Abstract
The gut microbiota (GM) has become recognized as a crucial element in preserving human fitness and influencing disease consequences. Commensal and pathogenic gut microorganisms are correlated with pathological progress in atherosclerosis (AS). GM may thus be a promising therapeutic target for AS. Natural products with cardioprotective qualities might improve the inflammation of AS by modulating the GM ecosystem, opening new avenues for researches and therapies. However, it is unclear what components of natural products are useful and what the actual mechanisms are. In this review, we have summarized the natural products relieving inflammation of AS by regulating the GM balance and active metabolites produced by GM.
Collapse
Affiliation(s)
- Bing Deng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liyu Tao
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiru Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Wu Z, Du Z, Tian Y, Liu M, Zhu K, Zhao Y, Wang H. Inulin accelerates weight loss in obese mice by regulating gut microbiota and serum metabolites. Front Nutr 2022; 9:980382. [PMID: 36245535 PMCID: PMC9554005 DOI: 10.3389/fnut.2022.980382] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Several studies indicated that the gut microbiota might participate in the beneficial effect of inulin on obesity. However, the mechanisms involved were still largely unknown. Sixteen high-fat diets (HFDs)-induced obese C57BL/6 mice were converted to a normal diet and then randomized into two groups, OND (obese mice + normal diet) group gavage-fed for 10 weeks with normal saline and ONDI (obese mice + normal diet + inulin) group with inulin at 10 g/kg/day. The body weight of HFD-induced obese mice showed different degrees of decrease in both groups. However, the ONDI group lost more weight and returned to normal earlier. Compared to the OND group, inulin supplementation significantly shifted the composition and structure of gut microbiota, such as higher α diversity. The β diversity analysis also confirmed the changes in gut microbiota composition between groups. At the genus level, the abundance of Alistipes was considerably increased, and it was significantly correlated with inulin supplementation (r = 0.72, P = 0.002). Serum metabolite levels were distinctly altered after inulin supplementation, and 143 metabolites were significantly altered in the ONDI group. Among them, indole-3-acrylic acid level increased more than 500-fold compared to the OND group. It was also strongly positive correlation with Alistipes (r = 0.72, P = 0.002) and inulin supplementation (r = 0.99, P = 9.2e−13) and negatively correlated with obesity (r = −0.72, P = 0.002). In conclusion, inulin supplementation could accelerate body weight loss in obese mice by increasing Alistipes and indole-3-acrylic acid level.
Collapse
Affiliation(s)
- Zeang Wu
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Zhenzhu Du
- Analysis and Testing Center, Shihezi University, Shihezi, China
| | - Yuanyuan Tian
- School of Medicine, Shihezi University, Shihezi, China
| | - Miao Liu
- School of Medicine, Shihezi University, Shihezi, China
| | - Kailong Zhu
- School of Medicine, Shihezi University, Shihezi, China
| | - Yufan Zhao
- School of Medicine, Shihezi University, Shihezi, China
| | - Haixia Wang
- School of Medicine, Shihezi University, Shihezi, China
- *Correspondence: Haixia Wang,
| |
Collapse
|