1
|
Munteanu C, Kotova P, Schwartz B. Impact of Olive Oil Components on the Expression of Genes Related to Type 2 Diabetes Mellitus. Nutrients 2025; 17:570. [PMID: 39940428 PMCID: PMC11820997 DOI: 10.3390/nu17030570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial metabolic disorder characterized by insulin resistance and beta cell dysfunction, resulting in hyperglycemia. Olive oil, a cornerstone of the Mediterranean diet, has attracted considerable attention due to its potential health benefits, including reducing the risk of developing T2DM. This literature review aims to critically examine and synthesize existing research regarding the impact of olive oil on the expression of genes relevant to T2DM. This paper also seeks to provide an immunological and genetic perspective on the signaling pathways of the main components of extra virgin olive oil. Key bioactive components of olive oil, such as oleic acid and phenolic compounds, were identified as modulators of insulin signaling. These compounds enhanced the insulin signaling pathway, improved lipid metabolism, and reduced oxidative stress by decreasing reactive oxygen species (ROS) production. Additionally, they were shown to alleviate inflammation by inhibiting the NF-κB pathway and downregulating pro-inflammatory cytokines and enzymes. Furthermore, these bioactive compounds were observed to mitigate endoplasmic reticulum (ER) stress by downregulating stress markers, thereby protecting beta cells from apoptosis and preserving their function. In summary, olive oil, particularly its bioactive constituents, has been demonstrated to enhance insulin sensitivity, protect beta cell function, and reduce inflammation and oxidative stress by modulating key genes involved in these processes. These findings underscore olive oil's therapeutic potential in managing T2DM. However, further research, including well-designed human clinical trials, is required to fully elucidate the role of olive oil in personalized nutrition strategies for the prevention and treatment of T2DM.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, 400372 Cluj-Napoca, Romania
| | - Polina Kotova
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 9190500, Israel
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 9190500, Israel
| |
Collapse
|
2
|
Arghavani H, Bilodeau JF, Rudkowska I. Association Between Circulating Fatty Acids and Blood Pressure: A Review. Curr Nutr Rep 2025; 14:15. [PMID: 39775363 DOI: 10.1007/s13668-024-00602-3] [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] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW High blood pressure (BP) or hypertension (HTN) remains key risk factors for cardiovascular disease (CVD). Circulating fatty acids (FAs) in the blood can affect directly cardiovascular hemodynamics and serves as building blocks for endocrine mediators modifying inflammatory processes and vascular function. This review aims to describe optimal circulating FA profiles for BP to adjust dietary recommendations for HTN prevention. RECENT FINDINGS Recent research highlights the critical role of FAs in regulating inflammation and vascular function. Different FAs have varying effects on oxidative stress, insulin resistance, inflammation, and vascular dysfunction, all contributing to HTN. These findings emphasize the importance of FAs in managing BP and preventing CVD. Up-to-now, findings suggest that eicosapentaenoic acid (20:5n3), docosahexaenoic acid (22:6n3), arachidic acid (20:0), behenic acid (22:0) and lignoceric acid (24:0) were promising candidates in reducing BP and thus, dietary intake could be recommended. Conversely, dietary intake of myristic acid (14:0), palmitic acid (16:0), and industrial trans FAs (iTFAs) should be restricted due to their association with elevated BP. Further research is warranted for pentadecanoic acid (15:0), heptadecanoic acid (17:0), stearic acid (18:0), alpha-linolenic acid (18:3n3), docosapentaenoic acid (22:5n3), linoleic acid (18:2n6), dihomo-gamma-linolenic acid (20:3n6), arachidonic acid (20:4n6), palmitoleic acid (16:1n7), and ruminant TFAs since their associations with BP present inconsistencies in the literature. Lifestyle factors such as dietary intake, physical activity, alcohol consumption and smoking should be considered when examining the relationship between FAs and BP. Overall, the FAs profile may contribute to BP level management; therefore, dietary recommendations are important.
Collapse
Affiliation(s)
- Hana Arghavani
- Endocrinology and Nephrology Research Axis, CHU de Québec Research Center, CHU of Quebec-Laval University, CHUL - 2705, Boulevard. Laurier, Quebec, G1V 4G2, Canada
| | - Jean-François Bilodeau
- Endocrinology and Nephrology Research Axis, CHU de Québec Research Center, CHU of Quebec-Laval University, CHUL - 2705, Boulevard. Laurier, Quebec, G1V 4G2, Canada
- Department of Medicine, Faculty of Medicine, Laval University, Quebec, QC, G1V 0A6, Canada
| | - Iwona Rudkowska
- Endocrinology and Nephrology Research Axis, CHU de Québec Research Center, CHU of Quebec-Laval University, CHUL - 2705, Boulevard. Laurier, Quebec, G1V 4G2, Canada.
- Department of Kinesiology, Faculty of Medicine, Laval University, Quebec, QC, G1V 0A6, Canada.
| |
Collapse
|
3
|
Tabashsum Z, Angeles-Solano M, Sidders AE, Parsons JB, Rowe SE. Palmitoleic acid sensitizes vancomycin-resistant Staphylococcus aureus to vancomycin by outpacing the expression of resistance genes. Microbiol Spectr 2025; 13:e0199624. [PMID: 39656010 PMCID: PMC11705785 DOI: 10.1128/spectrum.01996-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
The rise in antibiotic resistance limits the availability of antibiotics to treat bacterial infections. Despite this, antibiotic development pipelines remain sparse which makes using adjuvants to reverse antibiotic resistance a promising therapeutic strategy. The use of vancomycin, a frontline antibiotic used to treat hospitalized patients with methicillin-resistant Staphylococcus aureus (MRSA) infections, is complicated by high rates of treatment failure. Vancomycin binds to the D-ala-D-ala terminus of the nascent peptidoglycan precursor lipid II, preventing cell wall biosynthesis. Vancomycin-resistant strains of S. aureus and Enterococci typically express a van gene cluster that is induced in response to vancomycin and results in the synthesis of an alternative lipid precursor with a peptide chain ending in D-ala-D-lac. Vancomycin has low affinity for the D-ala-D-lac terminus, and the bacteria can resume growth even in the presence of an otherwise lethal dose of vancomycin. We previously showed that palmitoleic acid, a host-produced monounsaturated fatty acid, combined with vancomycin led to an accumulation of large fluid patches in the bacterial membrane, resulting in membrane destabilization and cell death. In this study, we observed that palmitoleic acid increases the rate of vancomycin killing by more than 50-fold, compared to vancomycin alone. This rapid bactericidal activity by the combined treatment sensitized vancomycin-resistant S. aureus (VRSA) and vancomycin-resistant Enterococcus (VRE) to vancomycin, likely by outpacing the expression of vancomycin resistance genes. This study represents an important step in the ongoing effort to mitigate antibiotic resistance.IMPORTANCEThe development of antibiotics has transformed medicine, reducing the incidence and severity of bacterial infections and allowing for advancements in healthcare, including invasive surgeries and organ transplants. However, the rise of antibiotic resistance poses a significant threat to these medical advancements, leading to treatment failures that result in increased patient morbidity and mortality, as well as substantial healthcare costs. Vancomycin-resistant Enterococcus (VRE) species are prevalent in hospital settings and chronic infections. Although high-level vancomycin resistance in S. aureus is rare, S. aureus can acquire plasmids expressing vancomycin resistance genes from resistant Enterococcal species during infection, further complicating treatment. In this study, we find that palmitoleic acid increases the rate of vancomycin killing and restores sensitivity to vancomycin-resistant S. aureus (VRSA) and VRE isolates.
Collapse
Affiliation(s)
- Zajeba Tabashsum
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michelle Angeles-Solano
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ashelyn E. Sidders
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joshua B. Parsons
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
4
|
Mensah EO, Danyo EK, Asase RV. Exploring the effect of different diet types on ageing and age-related diseases. Nutrition 2025; 129:112596. [PMID: 39488864 DOI: 10.1016/j.nut.2024.112596] [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: 03/15/2024] [Revised: 08/21/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024]
Abstract
In recent times, there has been growing interest in understanding the factors contributing to prolonged and healthy lifespans observed in specific populations, tribes, or countries. Factors such as environmental and dietary play significant roles in shaping the ageing process and are often the focus of inquiries seeking to unravel the secrets behind longevity. Among these factors, diet emerges as a primary determinant, capable of either promoting or mitigating the onset of age-related diseases that impact the ageing trajectory. This review examines the impact of various diet types on ageing and age-related conditions, including cardiovascular disease, cancer, neurodegenerative disorders, and metabolic syndrome. Different dietary patterns, such as the Mediterranean diet, the Japanese diet, vegetarian and vegan diets, as well as low-carbohydrate and ketogenic diets, are evaluated for their potential effects on longevity and health span. Each diet type is characterized by distinct nutritional profiles, emphasizing specific food groups, macronutrient compositions, and bioactive components, which may exert diverse effects on ageing processes and disease risk. Additionally, dietary factors such as calorie restriction, intermittent fasting, and dietary supplementation are explored for their potential anti-ageing and disease-modifying effects. Understanding the influence of various diet types on ageing and age-related diseases can inform personalized dietary recommendations and lifestyle interventions aimed at promoting healthy aging and mitigating age-associated morbidities.
Collapse
Affiliation(s)
- Emmanuel O Mensah
- Faculty of Ecotechnology, ITMO University, Saint Petersburg, Russian Federation.
| | - Emmanuel K Danyo
- Institute of Chemical Engineering, Ural Federal University, Yekaterinburg, Russian Federation
| | - Richard V Asase
- Institute of Chemical Engineering, Ural Federal University, Yekaterinburg, Russian Federation
| |
Collapse
|
5
|
Beldie LA, Dica CC, Moța M, Pirvu BF, Burticală MA, Mitrea A, Clenciu D, Efrem IC, Vladu BE, Timofticiuc DCP, Roșu MM, Gheonea TC, Amzolini AM, Moța E, Vladu IM. The Interactions Between Diet and Gut Microbiota in Preventing Gestational Diabetes Mellitus: A Narrative Review. Nutrients 2024; 16:4131. [PMID: 39683525 DOI: 10.3390/nu16234131] [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: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Recent studies have revealed that dysbiosis, defined as alterations in gut microbiota, plays an important role in the development and the progression of many non-communicable diseases, including metabolic disorders, such as type 2 diabetes mellitus and gestational diabetes mellitus (GDM). The high frequency of GDM makes this disorder an important public health issue, which needs to be addressed in order to reduce both the maternal and fetal complications that are frequently associated with this disease. The studies regarding the connections between gut dysbiosis and GDM are still in their early days, with new research continuously emerging. This narrative review seeks to outline the mechanisms through which a healthy diet that protects the gut microbiota is able to prevent the occurrence of GDM, thus providing medical nutritional therapeutic perspectives for the management of GDM.
Collapse
Affiliation(s)
- Luiza-Andreea Beldie
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Cristina-Camelia Dica
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Maria Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Bianca-Florentina Pirvu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Marilena-Alexandra Burticală
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Cristian Efrem
- Department of Medical Semiology, Faculty of Dentistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Beatrice Elena Vladu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Cristina Protasiewicz Timofticiuc
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Magdalena Roșu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Theodora Claudia Gheonea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anca Maria Amzolini
- Department of Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
6
|
Chen M, Wu Y, Yang H, Liu T, Han T, Dai W, Cen J, Ouyang F, Chen J, Liu J, Zhou L, Hu X. Effects of fermented Arctium lappa L. root by Lactobacillus casei on hyperlipidemic mice. Front Pharmacol 2024; 15:1447077. [PMID: 39529876 PMCID: PMC11551023 DOI: 10.3389/fphar.2024.1447077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction This study aimed to establish a fermentation system based on Lactobacillus casei (LC) and Arctium lappa L. root (AR) to investigate its effects. The objectives included comparing metabolite profiles pre- and post-fermentation using untargeted metabolomics and evaluating the impact of LC-AR in high-fat diet-induced hyperlipidemic mice. Methods Untargeted metabolomics was used to analyze differences in metabolites before and after fermentation. In vitro antioxidant activity, liver injury, lipid levels, pro-inflammatory cytokine levels, and cholesterol-related mRNA expression were assessed. 16S rRNA sequencing was conducted to evaluate changes in gut microbiota composition. Results LC-AR exhibited stronger antioxidant activity and higher metabolite levels than AR. It also improved liver injury as well as better regulation of lipid levels, pro-inflammatory cytokine levels, and cholesterol-related mRNA. 16S rRNA analysis revealed that LC-AR decreased the Firmicutes/Bacteroidetes ratio, which correlated negatively with triglycerides, total cholesterol, and low-density lipoprotein cholesterol levels. Discussion These findings suggest that LC-AR may serve as a promising functional food and drug raw material for improving hyperlipidemia, particularly through its beneficial effects on gut microbiota and lipid regulation.
Collapse
Affiliation(s)
- MingJu Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yuxiao Wu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Hongxuan Yang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Tianfeng Liu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Tongkun Han
- Shenzhen Bao’an District Songgang People’s Hospital, Shenzhen, Guangdong, China
| | - Wangqiang Dai
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Junyue Cen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Fan Ouyang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jingjing Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jianxin Liu
- School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua, Hunan, China
| | - Lin Zhou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xuguang Hu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Tang R, Zhu Y, Chen L, Tong J, Ma X, Sun F, Zheng L, Yu H, Yang J. Lipid metabolites abnormally expressed in pelvic fluid as potential biomarkers for ovarian cancer: A case-control study. J Proteomics 2024; 307:105261. [PMID: 39032862 DOI: 10.1016/j.jprot.2024.105261] [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: 03/12/2024] [Revised: 06/27/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Ovarian cancer is insidious and usually detected in advanced stages of the disease. As the ovaries are pelvic organs, changes in their pelvic fluid metabolites may be associated with ovarian cancer. METHODS Metabolomic changes in the pelvic fluid were detected using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in patients with ovarian cancer, ovarian cysts and uterine fibroids. Area under the curve (AUC) analysis was used to assess the diagnostic performance of lipid metabolites and blood tumor indices. The Pearson correlation algorithm was used to analyze the correlation between clinical characteristics and lipid metabolites in ovarian cancer patients. RESULTS There were 24 lipid metabolites significantly changed in the pelvic fluid of ovarian cancer patients (p < 0.05). Palmitoylcarnitine, lipoamide, lipid metabolites, and blood tumor indices (CA15-3 and CA125) showed AUC > 0.8, with palmitoylcarnitine reaching a high of 0.942. In addition, we found that some lipid metabolites were significantly associated with the clinical stage, abdominal water volume, lymphatic metastasis, and recurrence (p < 0.05, r > 0.5). CONCLUSION Levels of specific lipid metabolites are potential biomarkers of ovarian cancer and may play a key role in the early diagnosis and prognostic assessment of ovarian cancer. SIGNIFICANCE Our results showed that pelvic metabolites, especially some lipid metabolites, play an important role in the diagnosis of ovarian cancer. Meanwhile, partial lipid metabolites were closely associated with the clinical presentation and prognosis of patients with ovarian cancer. We believe that our study makes a significant contribution to the literature because it provides a potential approach that is more effective for ovarian cancer detection.
Collapse
Affiliation(s)
- Rongrong Tang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China; School of Medicine, ShaoXing University, ShaoXing City, Zhejiang Province, China
| | - Yunshan Zhu
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, China
| | - Lingfeng Chen
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, China
| | - Jinfei Tong
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, China
| | - Xudong Ma
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China
| | - Fangying Sun
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China; School of Medicine, ShaoXing University, ShaoXing City, Zhejiang Province, China
| | - Limei Zheng
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China
| | - Hailan Yu
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China
| | - Jianhua Yang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, PR China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, China.
| |
Collapse
|
8
|
Zhang Y, Li Y, Han Z, Huo Q, Ji L, Liu X, Li H, Zhu X, Hao Z. miR-328-5p functions as a critical negative regulator in early endothelial inflammation and advanced atherosclerosis. BMB Rep 2024; 57:375-380. [PMID: 38919016 PMCID: PMC11362139] [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: 04/10/2024] [Revised: 04/26/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Early proatherogenic inflammation constitutes a significant risk factor for atherogenesis development. Despite this, the precise molecular mechanisms driving this pathological progression largely remain elusive. Our study unveils a pivotal role for the microRNA miR-328-5p in dampening endothelial inflammation by modulating the stability of JUNB (JunB proto-oncogene). Perturbation of miR-328-5p levels results in heightened monocyte adhesion to endothelial cells and enhanced transendothelial migration, while its overexpression mitigates these inflammatory processes. Furthermore, miR-328-5p hinders macrophage polarization toward the pro-inflammatory M1 phenotype, and exerts a negative influence on atherosclerotic plaque formation in vivo. By pinpointing JUNB as a direct miR-328-5p target, our research underscores the potential of miR-328-5p as a therapeutic target for inflammatory atherosclerosis. Reintroduction of JUNB effectively counteracts the anti-atherosclerotic effects of miR-328-5p, highlighting the promise of pharmacological miR-328-5p targeting in managing inflammatory atherosclerosis. [BMB Reports 2024; 57(8): 375-380].
Collapse
Affiliation(s)
- Yangxia Zhang
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Yingke Li
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhisheng Han
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Qingyang Huo
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Longkai Ji
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Xuejia Liu
- Stem Cells and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Han Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Xinxing Zhu
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
- Department of Respiratory and Critical Care Medicine, Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, First Affiliated Hospital, Bengbu Medical University, Bengbu 233004, China
| | - Zhipeng Hao
- Department of Thoracic Surgery of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| |
Collapse
|
9
|
Won YS, Bak SG, Chandimali N, Park EH, Lim HJ, Kwon HS, Park SI, Lee SJ. 7-MEGA™ inhibits adipogenesis in 3T3-L1 adipocytes and suppresses obesity in high-fat-diet-induced obese C57BL/6 mice. Lipids Health Dis 2024; 23:192. [PMID: 38909257 PMCID: PMC11193219 DOI: 10.1186/s12944-024-02175-0] [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: 03/18/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Overweight, often known as obesity, is the abnormal and excessive accumulation of fat that exposes the health of a person at risk by increasing the likelihood that they may experience many chronic conditions. Consequently, obesity has become a global health threat, presenting serious health issues, and attracting a lot of attention in the healthcare profession and the scientific community. METHOD This study aims to explore the anti-adipogenic properties of 7-MEGA™ in an attempt to address obesity, using both in vitro and in vivo research. The effects of 7MEGA™ at three distinct concentrations were investigated in obese mice who were given a high-fat diet (HFD) and 3T3-L1 adipocytes. RESULTS 7MEGA™ decreased the total fat mass, overall body weight, and the perirenal and subcutaneous white adipose tissue (PWAT and SWAT) contents in HFD mice. Additionally, 7MEGA™ showed promise in improving the metabolic health of individuals with obesity and regulate the levels of insulin hormone, pro-inflammatory cytokines and adipokines. Furthermore, Peroxisome proliferator-activated receptors (PPAR) α and γ, Uncoupling Protein 1 (UCP-1), Sterol Regulatory Element-Binding Protein 1 (SREBP-1), Fatty Acid-Binding Protein 4 (FABP4), Fatty Acid Synthase (FAS), Acetyl-CoA Carboxylase (ACC), Stearoyl-CoA Desaturase-1 (SCD-1) and CCAAT/Enhancer-Binding Protein (C/EBPα) were among the adipogenic regulators that 7MEGA™ could regulate. CONCLUSION In summary, this study uncovered that 7MEGA™ demonstrates anti-adipogenic and anti-obesity effects, suggesting its potential in combating obesity.
Collapse
Affiliation(s)
- Yeong-Seon Won
- Division of Research Management, Department of Bioresource Industrialization, Honam National Institute of Biological Resource, Mokpo, 58762, Republic of Korea
| | - Seon-Gyeong Bak
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
| | - Nisansala Chandimali
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
- Department of Applied Biotechnology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Eun Hyun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyung-Jin Lim
- Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
| | - Hyuck Se Kwon
- R&D Team, Food & Supplement Health Claims, Vitech, Jeonju, 55365, Republic of Korea
| | - Sang-Ik Park
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Seung Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea.
- Department of Applied Biotechnology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
10
|
Li T, Hu X, Fan L, Yang Y, He K. Myricanol improves metabolic profiles in dexamethasone induced lipid and protein metabolism disorders in mice. Biomed Pharmacother 2024; 174:116557. [PMID: 38583337 DOI: 10.1016/j.biopha.2024.116557] [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: 12/15/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024] Open
Abstract
Myricanol (MY) is one of the main active components from bark of Myrica Rubra. It is demonstrated that MY rescues dexamethasone (DEX)-induced muscle dysfunction via activating silent information regulator 1 (SIRT1) and increasing adenosine 5'-monophosphate-activated protein kinase (AMPK) phosphorylation. Since SIRT1 and AMPK are widely involved in the metabolism of nutrients, we speculated that MY may exert beneficial effects on DEX-induced metabolic disorders. This study for the first time applied widely targeted metabolomics to investigate the beneficial effects of MY on glucose, lipids, and protein metabolism in DEX-induced metabolic abnormality in mice. The results showed that MY significantly reversed DEX-induced soleus and gastrocnemius muscle weight loss, muscle fiber damage, and muscle strength loss. MY alleviated DEX-induced metabolic disorders by increasing SIRT1 and glucose transporter type 4 (GLUT4) expressions. Additionally, myricanol prevented muscle cell apoptosis and atrophy by inhibiting caspase 3 cleavages and muscle ring-finger protein-1 (MuRF1) expression. Metabolomics showed that MY treatment reversed the serum content of carnitine ph-C1, palmitoleic acid, PS (16:0_17:0), PC (14:0_20:5), PE (P-18:1_16:1), Cer (t18:2/38:1(2OH)), four amino acids and their metabolites, and 16 glycerolipids in DEX mice. Kyoto encyclopedia of genes and genomes (KEGG) and metabolic set enrichment analysis (MSEA) analysis revealed that MY mainly affected metabolic pathways, glycerolipid metabolism, lipolysis, fat digestion and absorption, lipid and atherosclerosis, and cholesterol metabolism pathways through regulation of metabolites involved in glutathione, butanoate, vitamin B6, glycine, serine and threonine, arachidonic acid, and riboflavin metabolism. Collectively, MY can be used as an attractive therapeutic agent for DEX-induced metabolic abnormalities.
Collapse
Affiliation(s)
- Tiandan Li
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Xiaochao Hu
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Lingyang Fan
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Yong Yang
- chool of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, China.
| | - Kai He
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China.
| |
Collapse
|
11
|
Du H, Li D, Molive LM, Wu N. Advances in free fatty acid profiles in gestational diabetes mellitus. J Transl Med 2024; 22:180. [PMID: 38374136 PMCID: PMC10875910 DOI: 10.1186/s12967-024-04922-4] [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: 11/26/2023] [Accepted: 01/21/2024] [Indexed: 02/21/2024] Open
Abstract
The morbidity of gestational diabetes mellitus (GDM) is increasing and is associated with adverse perinatal outcomes and long-term maternal and infant health. The exact mechanism underlying changes in plasma free fatty acid (FFA) profiles in patients with GDM is unknown. However, it is believed that changes in diet and lipid metabolism may play a role. Fatty acids contain many specific FFAs, and the type of FFA has different impacts on physiological processes; hence, determining changes in FFAs in individual plasma is essential. Alterations in FFA concentration or profile may facilitate insulin resistance. Additionally, some FFAs show potential to predict GDM in early pregnancy and are strongly associated with the growth and development of the fetus and occurrence of macrosomia. Here, we aimed to review changes in FFAs in women with GDM and discuss the relationship of FFAs with GDM incidence and adverse outcomes.
Collapse
Affiliation(s)
- Haoyi Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Danyang Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Laura Monjowa Molive
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Na Wu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
- Medical Department, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang, People's Republic of China.
| |
Collapse
|
12
|
Cetin E, Pedersen B, Porter LM, Adler GK, Burak MF. Protocol for a randomized placebo-controlled clinical trial using pure palmitoleic acid to ameliorate insulin resistance and lipogenesis in overweight and obese subjects with prediabetes. Front Endocrinol (Lausanne) 2024; 14:1306528. [PMID: 38313838 PMCID: PMC10835623 DOI: 10.3389/fendo.2023.1306528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/27/2023] [Indexed: 02/06/2024] Open
Abstract
Palmitoleic acid (POA), a nonessential, monounsaturated omega-7 fatty acid (C16:1n7), is a lipid hormone secreted from adipose tissue and has beneficial effects on distant organs, such as the liver and muscle. Interestingly, POA decreases lipogenesis in toxic storage sites such as the liver and muscle, and paradoxically increases lipogenesis in safe storage sites, such as adipose tissue. Furthermore, higher POA levels in humans are correlated with better insulin sensitivity, an improved lipid profile, and a lower incidence of type-2 diabetes and cardiovascular pathologies, such as myocardial infarction. In preclinical animal models, POA improves glucose intolerance, dyslipidemia, and steatosis of the muscle and liver, while improving insulin sensitivity and secretion. This double-blind placebo-controlled clinical trial tests the hypothesis that POA increases insulin sensitivity and decreases hepatic lipogenesis in overweight and obese adult subjects with pre-diabetes. Important to note, that this is the first study ever to use pure (>90%) POA with < 0.3% palmitic acid (PA), which masks the beneficial effects of POA. The possible positive findings may offer a therapeutic and/or preventative pathway against diabetes and related immunometabolic diseases.
Collapse
Affiliation(s)
- Ecesu Cetin
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Brian Pedersen
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Lindsey M. Porter
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Gail K. Adler
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Mehmet Furkan Burak
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Sabri Ulker Center, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| |
Collapse
|
13
|
Yu Q, Yang Y, Xu T, Cai Y, Yang Z, Yuan F. Palmitoleic acid protects microglia from palmitate-induced neurotoxicity in vitro. PLoS One 2024; 19:e0297031. [PMID: 38241239 PMCID: PMC10798504 DOI: 10.1371/journal.pone.0297031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/26/2023] [Indexed: 01/21/2024] Open
Abstract
Although palmitoleic acid (POA) is a lipokine with beneficial effects on obesity and is produced as a byproduct from the manufacture of prescription omega-3 fatty acids, its role in nervous system inflammation is still unknown. This study aims to examine the mechanisms and protective effects of POA against palmitic acid (PA)-induced microglial death. PA-induced microglial death was used as a model for POA intervention. Various inhibitors were employed to suppress potential routes of PA entry into the cell. Immunofluorescence staining and Western blotting were conducted to elucidate the protective pathways involved. The results suggest POA has the potential to eliminate PA-induced lactate dehydrogenase (LDH) release, which decreases the overall number of propidium iodide (PI)-positive cells compared with control. Moreover, POA has the potential to significantly increase lipid droplets (LDs) in the cytoplasm, without causing any lysosomal damage. POA inhibited both canonical and non-canonical gasdermin D (GSDMD)-mediated pyroptosis and gasdermin E (GSDME)-mediated pyroptosis, which PA typically induces. Additionally, POA inhibited the endoplasmic reticulum (ER) stress and apoptosis-related proteins induced by PA. Based on the findings, POA can exert a protective effect on microglial death induced by PA via pathways related to pyroptosis, apoptosis, ER stress, and LDs.
Collapse
Affiliation(s)
- Qingting Yu
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Yanzhuo Yang
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Ting Xu
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | | | - Zuisu Yang
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Falei Yuan
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| |
Collapse
|
14
|
Chen Y, Li Y, Fan Y, Chen S, Chen L, Chen Y, Chen Y. Gut microbiota-driven metabolic alterations reveal gut-brain communication in Alzheimer's disease model mice. Gut Microbes 2024; 16:2302310. [PMID: 38261437 PMCID: PMC10807476 DOI: 10.1080/19490976.2024.2302310] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/03/2024] [Indexed: 01/25/2024] Open
Abstract
The gut microbiota (GM) and its metabolites affect the host nervous system and are involved in the pathogeneses of various neurological diseases. However, the specific GM alterations under pathogenetic pressure and their contributions to the "microbiota - metabolite - brain axis" in Alzheimer's disease (AD) remain unclear. Here, we investigated the GM and the fecal, serum, cortical metabolomes in APP/PS1 and wild-type (WT) mice, revealing distinct hub bacteria in AD mice within scale-free GM networks shared by both groups. Moreover, we identified diverse peripheral - central metabolic landscapes between AD and WT mice that featured bile acids (e.g. deoxycholic and isodeoxycholic acid) and unsaturated fatty acids (e.g. 11Z-eicosenoic and palmitoleic acid). Machine-learning models revealed the relationships between the differential/hub bacteria and these metabolic signatures from the periphery to the brain. Notably, AD-enriched Dubosiella affected AD occurrence via cortical palmitoleic acid and vice versa. Considering the transgenic background of the AD mice, we propose that Dubosiella enrichment impedes AD progression via the synthesis of palmitoleic acid, which has protective properties against inflammation and metabolic disorders. We identified another association involving fecal deoxycholic acid-mediated interactions between the AD hub bacteria Erysipelatoclostridium and AD occurrence, which was corroborated by the correlation between deoxycholate levels and cognitive scores in humans. Overall, this study elucidated the GM network alterations, contributions of the GM to peripheral - central metabolic landscapes, and mediatory roles of metabolites between the GM and AD occurrence, thus revealing the critical roles of bacteria in AD pathogenesis and gut - brain communications under pathogenetic pressure.
Collapse
Affiliation(s)
- Yijing Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yinhu Li
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yingying Fan
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Shuai Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Li Chen
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
15
|
Sun Q, Xing X, Wang H, Wan K, Fan R, Liu C, Wang Y, Wu W, Wang Y, Wang R. SCD1 is the critical signaling hub to mediate metabolic diseases: Mechanism and the development of its inhibitors. Biomed Pharmacother 2024; 170:115586. [PMID: 38042113 DOI: 10.1016/j.biopha.2023.115586] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 12/04/2023] Open
Abstract
Metabolic diseases, featured with dysregulated energy homeostasis, have become major global health challenges. Patients with metabolic diseases have high probability to manifest multiple complications in lipid metabolism, e.g. obesity, insulin resistance and fatty liver. Therefore, targeting the hub genes in lipid metabolism may systemically ameliorate the metabolic diseases, along with the complications. Stearoyl-CoA desaturase 1(SCD1) is a key enzyme that desaturates the saturated fatty acids (SFAs) derived from de novo lipogenesis or diet to generate monounsaturated fatty acids (MUFAs). SCD1 maintains the metabolic and tissue homeostasis by responding to, and integrating the multiple layers of endogenous stimuli, which is mediated by the synthesized MUFAs. It critically regulates a myriad of physiological processes, including energy homeostasis, development, autophagy, tumorigenesis and inflammation. Aberrant transcriptional and epigenetic activation of SCD1 regulates AMPK/ACC, SIRT1/PGC1α, NcDase/Wnt, etc, and causes aberrant lipid accumulation, thereby promoting the progression of obesity, non-alcoholic fatty liver, diabetes and cancer. This review critically assesses the integrative mechanisms of the (patho)physiological functions of SCD1 in metabolic homeostasis, inflammation and autophagy. For translational perspective, potent SCD1 inhibitors have been developed to treat various types of cancer. We thus discuss the multidisciplinary advances that greatly accelerate the development of SCD1 new inhibitors. In conclusion, besides cancer treatment, SCD1 may serve as the promising target to combat multiple metabolic complications simultaneously.
Collapse
Affiliation(s)
- Qin Sun
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaorui Xing
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Huanyu Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Kang Wan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Ruobing Fan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Cheng Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yongjian Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Wenyi Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
16
|
Zhan Q, Thakur K, Feng JY, Zhu YY, Zhang JG, Wei ZJ. LC-MS based metabolomics analysis of okara fermented by Bacillus subtilis DC-15: Insights into nutritional and functional profile. Food Chem 2023; 413:135656. [PMID: 36780856 DOI: 10.1016/j.foodchem.2023.135656] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
Recent studies emphasize the improved nutritional and functional status of fermented okara; however, little is known about the metabolite change during fermentation and how it alters metabolic pathways. A metabolomics approach based on untargeted LC-MS reveals metabolic changes in okara fermented by Bacillus subtilis DC-15. We identified 761 differential metabolites, with the highest abundances found in amino acids, dipeptides, fatty acids, small molecule sugars, and vitamins. Moreover, these identified metabolites were mapped to their respective biosynthesis pathways in order to gain a better understanding of the biochemical reactions triggered by fermentation. Based on Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, 485 metabolites were enriched to metabolism-related pathways. They include 37 carbohydrate metabolites, 79 amino acid metabolites, and 22 lipid metabolites. As a result of okara fermentation, we observed a gradual enrichment of metabolites and stabilization of the compounds.
Collapse
Affiliation(s)
- Qi Zhan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China; Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, School of Biological Science and Engineering, North Minzu University, Yinchuan 750021, People's Republic of China.
| | - Kiran Thakur
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China; Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, School of Biological Science and Engineering, North Minzu University, Yinchuan 750021, People's Republic of China.
| | - Jing-Yu Feng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China.
| | - Yun-Yang Zhu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China.
| | - Jian-Guo Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China; Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, School of Biological Science and Engineering, North Minzu University, Yinchuan 750021, People's Republic of China.
| | - Zhao-Jun Wei
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China; Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, School of Biological Science and Engineering, North Minzu University, Yinchuan 750021, People's Republic of China.
| |
Collapse
|
17
|
Fatty acids act on vascular endothelial cells and influence the development of cardiovascular disease. Prostaglandins Other Lipid Mediat 2023; 165:106704. [PMID: 36621562 DOI: 10.1016/j.prostaglandins.2023.106704] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Endothelial cells (ECs) maintain the health of blood vessels and prevent the development of cardiovascular disease (CVD). Free saturated fatty acids (FAs) induce EC damage and increase the risk of CVD by promoting arteriosclerosis. Conversely, polyunsaturated FAs (PUFAs), such as docosahexaenoic acid, are thought to suppress EC damage induced during the early stages of CVD. This review describes the effects of multiple dietary FAs on EC disorders involved in the development of CVD. The roles of FAs in atherosclerosis and CVD were analyzed by evaluating articles published in PubMed, Science Direct, and Web of Science. Saturated FAs were found to induce EC damage by reducing the production and action of EC-derived nitric oxide. Oxidative stress, inflammation, and the renin-angiotensin system were found to be involved in EC disorder. Furthermore, n-3 PUFAs were found to reduce EC dysfunction and prevent the development of EC disorder. These results indicate that FAs may affect EC failure induced during the early stages of CVD and reduce the risk of developing the disease.
Collapse
|
18
|
Goffin N, Buache E, Charpentier C, Lehrter V, Morjani H, Gobinet C, Piot O. Trajectory Inference for Unraveling Dynamic Biological Processes from Raman Spectral Data. Anal Chem 2023; 95:4395-4403. [PMID: 36788139 DOI: 10.1021/acs.analchem.2c04901] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Cell heterogeneity is a crucial parameter for understanding the complexity of numerous biomedical issues. Trajectory inference-based approaches are recent tools developed for single-cell transcriptomics (scRNA-seq) data analysis. They aim to reconstruct evolving pathways from the variety of cell states that coexist simultaneously in a cell population. We propose to expand this concept to Raman spectroscopy, a label-free modality that probes the global molecular nature of a sample, by investigating the dynamics of adipocyte differentiation.
Collapse
Affiliation(s)
- Nicolas Goffin
- University of Reims Champagne Ardenne, BioSpecT EA 7506, SFR Santé, France
| | - Emilie Buache
- University of Reims Champagne Ardenne, BioSpecT EA 7506, SFR Santé, France
| | - Celine Charpentier
- University of Reims Champagne Ardenne, BioSpecT EA 7506, SFR Santé, France
| | - Véronique Lehrter
- University of Reims Champagne Ardenne, BioSpecT EA 7506, SFR Santé, France
| | - Hamid Morjani
- University of Reims Champagne Ardenne, BioSpecT EA 7506, SFR Santé, France
| | - Cyril Gobinet
- University of Reims Champagne Ardenne, BioSpecT EA 7506, SFR Santé, France
| | - Olivier Piot
- University of Reims Champagne Ardenne, BioSpecT EA 7506, SFR Santé, France.,University of Reims Champagne Ardenne, Platform of Cellular and Tissular Imaging (PICT) EA 7506, SFR Santé, France
| |
Collapse
|
19
|
Salsinha AS, Rodríguez-Alcalá LM, Pimentel LL, Pintado M. Role of bioactive lipids in obesity. BIOACTIVE LIPIDS 2023:133-167. [DOI: 10.1016/b978-0-12-824043-4.00012-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Sun Y, Hao L, Han W, Luo J, Zheng J, Yuan D, Ye H, Li Q, Huang G, Han T, Yang Z. Intrafollicular fluid metabolic abnormalities in relation to ovarian hyperstimulation syndrome: Follicular fluid metabolomics via gas chromatography-mass spectrometry. Clin Chim Acta 2023; 538:189-202. [PMID: 36566958 DOI: 10.1016/j.cca.2022.11.033] [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: 10/23/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Ovarian hyperstimulation syndrome (OHSS) is the most serious iatrogenic complication of ovulation stimulation during assisted reproductive technology. The main objective of this study was to investigate intrafollicular fluid metabolic change profiles of OHSS in non-ovarian etiologic infertility women (CON) and polycystic ovarian syndrome patients (PCOS). METHODS 87 infertile women were divided into four subgroups: CON-Norm (CON with normal ovarian response), CON-OHSS (CON with OHSS), PCOS-Norm (PCOS with normal ovarian response), and PCOS-OHSS (PCOS with OHSS). The intrafollicular fluid metabolic profiles were analyzed with gas chromatography-mass spectrometry. The multivariable-adjusted conditional logistic regression was applied to assess the association of metabolites with OHSS risk. RESULTS We identified 17 and 3 metabolites that related to OHSS risk in CON and PCOS, respectively. 13 OHSS risk-related metabolites in CON were unsaturated fatty acids, 8 of which were also the significantly altered metabolites between all PCOS and CON-Norm. CONCLUSION Our study may shed light on the role of intrafollicular fluid metabolic abnormalities in the pathophysiology of OHSS. The findings suggested that there might be some metabolic heterogeneities underlying the development of OHSS in CON and PCOS women and indicated possible shared etiological factors in the development of PCOS and OHSS.
Collapse
Affiliation(s)
- Yixuan Sun
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R.China; Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R.China
| | - Lijuan Hao
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R.China
| | - Wei Han
- Center for Reproductive Medicine, Reproductive and Genetic Institute, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R.China
| | - Jing Luo
- Department of Pathology, Basic Medical College of Chongqing Medical University, 400016, P.R.China
| | - Jing Zheng
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R.China
| | - Dong Yuan
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R.China
| | - Hong Ye
- Center for Reproductive Medicine, Reproductive and Genetic Institute, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R.China
| | - Qinke Li
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R.China
| | - Guoning Huang
- Center for Reproductive Medicine, Reproductive and Genetic Institute, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R.China.
| | - Tingli Han
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R.China.
| | - Zhu Yang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R.China.
| |
Collapse
|
21
|
Muthuraj PG, Krishnamoorthy C, Anderson-Berry A, Hanson C, Natarajan SK. Novel Therapeutic Nutrients Molecules That Protect against Zika Virus Infection with a Special Note on Palmitoleate. Nutrients 2022; 15:124. [PMID: 36615782 PMCID: PMC9823984 DOI: 10.3390/nu15010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/11/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV) is a Flavivirus from the Flaviviridae family and a positive-sense single strand RNA virus. ZIKV infection can cause a mild infection to the mother but can be vertically transmitted to the developing fetus, causing congenital anomalies. The prevalence of ZIKV infections was relatively insignificant with sporadic outbreaks in the Asian and African continents until 2006. However, recent epidemic in the Caribbean showed significant increased incidence of Congenital Zika Syndrome. ZIKV infection results in placental pathology which plays a crucial role in disease transmission from mother to fetus. Currently, there is no Food and Drug Administration (FDA) approved vaccine or therapeutic drug against ZIKV. This review article summarizes the recent advances on ZIKV transmission and diagnosis and reviews nutraceuticals which can protect against the ZIKV infection. Further, we have reviewed recent advances related to the novel therapeutic nutrient molecules that have been shown to possess activity against Zika virus infected cells. We also review the mechanism of ZIKV-induced endoplasmic reticulum and apoptosis and the protective role of palmitoleate (nutrient molecule) against ZIKV-induced ER stress and apoptosis in the placental trophoblasts.
Collapse
Affiliation(s)
- Philma Glora Muthuraj
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Chandan Krishnamoorthy
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Ann Anderson-Berry
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Corrine Hanson
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Medical Nutrition Education, College of Allied Health Profession, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Medical Nutrition Education, College of Allied Health Profession, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
22
|
Roles of Palmitoleic Acid and Its Positional Isomers, Hypogeic and Sapienic Acids, in Inflammation, Metabolic Diseases and Cancer. Cells 2022; 11:cells11142146. [PMID: 35883589 PMCID: PMC9319324 DOI: 10.3390/cells11142146] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
In the last few years, the monounsaturated hexadecenoic fatty acids are being increasingly considered as biomarkers of health with key functions in physiology and pathophysiology. Palmitoleic acid (16:1n-7) and sapienic acid (16:1n-10) are synthesized from palmitic acid by the action of stearoyl-CoA desaturase-1 and fatty acid desaturase 2, respectively. A third positional isomer, hypogeic acid (16:1n-9) is produced from the partial β-oxidation of oleic acid. In this review, we discuss the current knowledge of the effects of palmitoleic acid and, where available, sapienic acid and hypogeic acid, on metabolic diseases such as diabetes, cardiovascular disease, and nonalcoholic fatty liver disease, and cancer. The results have shown diverse effects among studies in cell lines, animal models and humans. Palmitoleic acid was described as a lipokine able to regulate different metabolic processes such as an increase in insulin sensitivity in muscle, β cell proliferation, prevention of endoplasmic reticulum stress and lipogenic activity in white adipocytes. Numerous beneficial effects have been attributed to palmitoleic acid, both in mouse models and in cell lines. However, its role in humans is not fully understood, and is sometimes controversial. Regarding sapienic acid and hypogeic acid, studies on their biological effects are still scarce, but accumulating evidence suggests that they also play important roles in metabolic regulation. The multiplicity of effects reported for palmitoleic acid and the compartmentalized manner in which they often occur, may suggest the overlapping actions of multiple isomers being present at the same or neighboring locations.
Collapse
|
23
|
The Role of MicroRNAs in Hyperlipidemia: From Pathogenesis to Therapeutical Application. Mediators Inflamm 2022; 2022:3101900. [PMID: 35757107 PMCID: PMC9232323 DOI: 10.1155/2022/3101900] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
Hyperlipidemia is a common metabolic disorder with high morbidity and mortality, which brings heavy burden on social. Understanding its pathogenesis and finding its potential therapeutic targets are the focus of current research in this field. In recent years, an increasing number of studies have proved that miRNAs play vital roles in regulating lipid metabolism and were considered as promising therapeutic targets for hyperlipidemia and related diseases. It is demonstrated that miR-191, miR-222, miR-224, miR-27a, miR-378a-3p, miR-140-5p, miR-483, and miR-520d-5p were closely associated with the pathogenesis of hyperlipidemia. In this review, we provide brief overviews about advances in miRNAs in hyperlipidemia and its potential clinical application value.
Collapse
|
24
|
Concentrates from tender coconut water and coconut testa beneficially modulates tissue lipid profiles in high-fat fed rats. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2022; 59:1649-1657. [PMID: 35250088 PMCID: PMC8882753 DOI: 10.1007/s13197-021-05178-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 05/29/2021] [Accepted: 06/11/2021] [Indexed: 10/20/2022]
Abstract
The present study involved feeding of tender coconut water concentrate (TCW) and testa phenolic concentrate (PHE) to high fat fed animals. Analysis of serum showed increased HDL-C in concentrates treated group and therefore higher protection against atherogenesis compared to high fat fed group. Lesser fecal fat content and higher level of liver cholesterol were observed in high fat fed group, treatments with TCW and PHE ameliorated significantly. Fat content in the organs (liver, heart, kidney and adipose) of HFD group was high. Furthermore, certain fatty acids observed in the tissues were 14:0, 16:0, 18:0, 18:1, 18:2, 20:0, 20:3 and 20:4, among which 14:0, 16:0 & 18:0 are the major saturated fatty acids that increased significantly in high fat fed group when compared with starch fed group and ameliorated with TCW and PHE dose dependently. These results clearly indicate both the concentrates (TCW and PHE) restrict excess accumulation of lipids and altered composition of fatty acids in tissues as well as serum which are the risk factors for development of insulin resistance, hyperlipidemia, dyslipidemia etc., related to obesity. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13197-021-05178-2.
Collapse
|
25
|
Miklankova D, Markova I, Hüttl M, Stankova B, Malinska H. The Different Insulin-Sensitising and Anti-Inflammatory Effects of Palmitoleic Acid and Oleic Acid in a Prediabetes Model. J Diabetes Res 2022; 2022:4587907. [PMID: 36147256 PMCID: PMC9489414 DOI: 10.1155/2022/4587907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/01/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Monounsaturated fatty acids (MUFA) are understood to have therapeutic and preventive effects on chronic complications associated with type 2 diabetes mellitus (T2DM); however, there are differences between individual MUFAs. Although the effects of palmitoleic acid (POA) are still debated, POA can regulate glucose homeostasis, lipid metabolism, and cytokine production, thus improving metabolic disorders. In this study, we investigated and compared the metabolic effects of POA and oleic acid (OA) supplementation on glucose and lipid metabolism, insulin sensitivity, and inflammation in a prediabetic model, the hereditary hypertriglyceridemic rat (HHTg). HHTg rats exhibiting genetically determined hypertriglyceridemia, insulin resistance, and impaired glucose tolerance were fed a standard diet. POA and OA were each administered intragastrically at a dose of 100 mg/kg b.wt. for four weeks. RESULTS Supplementation with both MUFAs significantly elevated insulin and glucagon levels, but only POA decreased nonfasting glucose. POA-treated rats showed elevated circulating NEFA associated with increased lipolysis, lipoprotein lipase gene expression, and fatty acid reesterification in visceral adipose tissue (VAT). The mechanism of improved insulin sensitivity of peripheral tissues (measured as insulin-stimulated lipogenesis and glycogenesis) in POA-treated HHTg rats could contribute increased circulating adiponectin and omentin levels together with elevated FADS1 gene expression in VAT. POA-supplemented rats exhibited markedly decreased proinflammatory cytokine production by VAT, which can alleviate chronic inflammation. OA-supplemented rats exhibited decreased arachidonic acid (AA) profiles and decreased proinflammatory AA-derived metabolites (20-HETE) in membrane phospholipids of peripheral tissues. Slightly increased FADS1 gene expression after OA along with increased adiponectin production by VAT was reflected in slightly ameliorated adipose tissue insulin sensitivity (increased insulin-stimulated lipogenesis). CONCLUSIONS Our results show that POA served as a lipokine, ameliorating insulin sensitivity in peripheral tissue and markedly modulating the metabolic activity of VAT including cytokine secretion. OA had a beneficial effect on lipid metabolism and improved inflammation by modulating AA metabolism.
Collapse
Affiliation(s)
- Denisa Miklankova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Irena Markova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martina Hüttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Barbora Stankova
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Malinska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
26
|
Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Jukema JW. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol 2021; 18:824-837. [PMID: 34253911 DOI: 10.1038/s41569-021-00582-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
T cells are among the most common cell types present in atherosclerotic plaques and are increasingly being recognized as a central mediator in atherosclerosis development and progression. At the same time, triglycerides and fatty acids have re-emerged as crucial risk factors for atherosclerosis. Triglycerides and fatty acids are important components of the milieu to which the T cell is exposed from the circulation to the plaque, and increasing evidence shows that fatty acids influence T cell function. In this Review, we discuss the effects of fatty acids on four components of the T cell response - metabolism, activation, proliferation and polarization - and the influence of these changes on the pathogenesis of atherosclerosis. We also discuss how quiescent T cells can undergo a type of metabolic reprogramming induced by exposure to fatty acids in the circulation that influences the subsequent functions of T cells after activation, such as in atherosclerotic plaques.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands.
- Netherlands Heart Institute, Utrecht, Netherlands.
| |
Collapse
|
27
|
Ferroptosis: the potential value target in atherosclerosis. Cell Death Dis 2021; 12:782. [PMID: 34376636 PMCID: PMC8355346 DOI: 10.1038/s41419-021-04054-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/08/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022]
Abstract
In advanced atherosclerosis (AS), defective function-induced cell death leads to the formation of the characteristic necrotic core and vulnerable plaque. The forms and mechanisms of cell death in AS have recently been elucidated. Among them, ferroptosis, an iron-dependent form of necrosis that is characterized by oxidative damage to phospholipids, promotes AS by accelerating endothelial dysfunction in lipid peroxidation. Moreover, disordered intracellular iron causes damage to macrophages, vascular smooth muscle cells (VSMCs), vascular endothelial cells (VECs), and affects many risk factors or pathologic processes of AS such as disturbances in lipid peroxidation, oxidative stress, inflammation, and dyslipidemia. However, the mechanisms through which ferroptosis initiates the development and progression of AS have not been established. This review explains the possible correlations between AS and ferroptosis, and provides a reliable theoretical basis for future studies on its mechanism.
Collapse
|
28
|
Maternal diet high in linoleic acid alters offspring fatty acids and cardiovascular function in a rat model. Br J Nutr 2021; 127:540-553. [PMID: 33858529 DOI: 10.1017/s0007114521001276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Linoleic acid (LA), an essential n-6 fatty acid (FA), is critical for fetal development. We investigated the effects of maternal high LA (HLA) diet on offspring cardiac development and its relationship to circulating FA and cardiovascular function in adolescent offspring, and the ability of the postnatal diet to reverse any adverse effects. Female Wistar Kyoto rats were fed low LA (LLA; 1·44 % energy from LA) or high LA (HLA; 6·21 % energy from LA) diets for 10 weeks before pregnancy and during gestation/lactation. Offspring, weaned at postnatal day 25, were fed LLA or HLA diets and euthanised at postnatal day 40 (n 6-8). Maternal HLA diet decreased circulating total cholesterol and HDL-cholesterol in females and decreased total plasma n-3 FA in males, while maternal and postnatal HLA diets decreased total plasma n-3 FA in females. α-Linolenic acid (ALA) and EPA were decreased by postnatal but not maternal HLA diets in both sexes. Maternal and postnatal HLA diets increased total plasma n-6 and LA, and a maternal HLA diet increased circulating leptin, in both male and female offspring. Maternal HLA decreased slopes of systolic and diastolic pressure-volume relationship (PVR), and increased cardiac Col1a1, Col3a1, Atp2a1 and Notch1 in males. Maternal and postnatal HLA diets left-shifted the diastolic PVR in female offspring. Coronary reactivity was altered in females, with differential effects on flow repayment after occlusion. Thus, maternal HLA diets impact lipids, FA and cardiac function in offspring, with postnatal diet modifying FA and cardiac function in the female offspring.
Collapse
|
29
|
Monounsaturated Fatty Acids in Obesity-Related Inflammation. Int J Mol Sci 2020; 22:ijms22010330. [PMID: 33396940 PMCID: PMC7795523 DOI: 10.3390/ijms22010330] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/23/2020] [Accepted: 12/25/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is an important aspect of the metabolic syndrome and is often associated with chronic inflammation. In this context, inflammation of organs participating in energy homeostasis (such as liver, adipose tissue, muscle and pancreas) leads to the recruitment and activation of macrophages, which secrete pro-inflammatory cytokines. Interleukin-1β secretion, sustained C-reactive protein plasma levels and activation of the NLRP3 inflammasome characterize this inflammation. The Stearoyl-CoA desaturase-1 (SCD1) enzyme is a central regulator of lipid metabolism and fat storage. This enzyme catalyzes the generation of monounsaturated fatty acids (MUFAs)-major components of triglycerides stored in lipid droplets-from saturated fatty acid (SFA) substrates. In this review, we describe the molecular effects of specific classes of fatty acids (saturated and unsaturated) to better understand the impact of different diets (Western versus Mediterranean) on inflammation in a metabolic context. Given the beneficial effects of a MUFA-rich Mediterranean diet, we also present the most recent data on the role of SCD1 activity in the modulation of SFA-induced chronic inflammation.
Collapse
|
30
|
Determination of Major Endogenous FAHFAs in Healthy Human Circulation: The Correlations with Several Circulating Cardiovascular-Related Biomarkers and Anti-Inflammatory Effects on RAW 264.7 Cells. Biomolecules 2020; 10:biom10121689. [PMID: 33348748 PMCID: PMC7766943 DOI: 10.3390/biom10121689] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Fatty acid esters of hydroxy fatty acids (FAHFAs) are newly discovered long-chain fatty acids. However, the major endogenous FAHFAs in healthy human circulation, their correlation with cardiovascular (CV) biomarkers, and their anti-inflammatory effects have not been investigated and remain unclear. In the present study, a total of 57 healthy subjects were recruited. Liquid chromatography–mass spectrometry (LC-MS) was developed for the simultaneous determination of seven FAHFAs, four long-chain fatty acids, and four non-traditional circulating CV-related biomarkers. We found two major types of FAHFAs in healthy human circulation, palmitoleic acid ester of 9-hydroxystearic acid (9-POHSA), and oleic acid ester of 9-hydroxystearic acid (9-OAHSA). Both 9-POHSA and 9-OAHSA had a strong positive correlation with each other and were negatively correlated with fasting blood glucose, S-adenosyl-l-homocysteine (SAH), and trimethylamine N-oxide (TMAO), but not with l-homocysteine. 9-POHSA was also positively correlated with l-carnitine. Moreover, we confirmed that both 9-POHSA and 9-OAHSA exhibited an anti-inflammatory effect by suppressing LPS stimulated cytokines, including IL-1β and IL-6 in RAW 264.7 cells. In addition, palmitoleic acid also had a positive correlation with 9-POHSA and 9-OAHSA. As far as we know, this is the first report showing the major endogenous FAHFAs in healthy subjects and their CV protection potential which might be correlated with SAH and TMAO reduction, l-Carnitine elevation, and their anti-inflammatory effects.
Collapse
|
31
|
Scheja L, Heeren J. Novel Adipose Tissue Targets to Prevent and Treat Atherosclerosis. Handb Exp Pharmacol 2020; 270:289-310. [PMID: 33373032 DOI: 10.1007/164_2020_363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipose tissue as a major organ of lipid and lipoprotein metabolism has a major impact on metabolic homeostasis and thus influences the development of atherosclerosis and related cardiometabolic diseases. Unhealthy adipose tissue, which is often associated with obesity and systemic insulin resistance, promotes the development of diabetic dyslipidemia and can negatively affect vascular tissue homeostasis by secreting pro-inflammatory peptides and lipids. Conversely, paracrine and endocrine factors that are released from healthy adipose tissue can preserve metabolic balance and a functional vasculature. In this chapter, we describe adipose tissue types relevant for atherosclerosis and address the question how lipid metabolism as well as regulatory molecules produced in these fat depots can be targeted to counteract atherogenic processes in the vessel wall and improve plasma lipids. We discuss the role of adipose tissues in the action of approved drugs with anti-atherogenic activity. In addition, we present potential novel targets and therapeutic approaches aimed at increasing lipoprotein disposal in adipose tissue, boosting the activity of heat-producing (thermogenic) adipocytes, reducing adipose tissue inflammation, and improving or replacing beneficial hormones released from adipose tissues. Furthermore, we describe the future potential of innovative drug delivery technologies.
Collapse
Affiliation(s)
- Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
32
|
Huang NK, Matthan NR, Galluccio JM, Shi P, Lichtenstein AH, Mozaffarian D. Supplementation with Seabuckthorn Oil Augmented in 16:1n-7t Increases Serum Trans-Palmitoleic Acid in Metabolically Healthy Adults: A Randomized Crossover Dose-Escalation Study. J Nutr 2020; 150:1388-1396. [PMID: 32140719 PMCID: PMC7269729 DOI: 10.1093/jn/nxaa060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/20/2019] [Accepted: 02/24/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In animal models cis-palmitoleic acid (9-hexadecenoic acid; 16:1n-7c), a lipokine, improves insulin sensitivity, inflammation, and lipoprotein profiles; in humans trans-palmitoleic acid (16:1n-7t) has been associated with lower incidence of type 2 diabetes. The response to dose-escalation of supplements containing cis- and trans-palmitoleic acid has not been evaluated. OBJECTIVES We examined dose-escalation effects of oral supplementation with seabuckthorn oil and seabuckthorn oil augmented in 16:1n-7t on serum phospholipid fatty acids (PLFAs). METHODS Thirteen participants (7 women and 6 men; age 48 ± 16 y, BMI 30.4 ± 3.7 kg/m2) participated in a randomized, double-blind, crossover, dose-escalation trial of unmodified seabuckthorn oils relatively high in 16:1n-7c (380, 760, and 1520 mg 16:1n-7c/d) and seabuckthorn oils augmented in 16:1n-7t (120, 240, and 480 mg 16:1n-7t/d). Each of the 3 escalation doses was provided for 3 wk, with a 4-wk washout period between the 2 supplements. At the end of each dose period, fasting blood samples were used to determine the primary outcomes (serum concentrations of the PLFAs 16:1n-7t and 16:1n-7c) and the secondary outcomes (glucose homeostasis, serum lipids, and clinical measures). Trends across doses were evaluated using linear regression. RESULTS Compared with baseline, supplementation with seabuckthorn oil augmented in 16:1n-7t increased phospholipid 16:1n-7t by 26.6% at the highest dose (P = 0.0343). Supplementation with unmodified seabuckthorn oil resulted in a positive trend across the dose-escalations (P-trend = 0.0199). No significant effects of either supplement were identified on blood glucose, insulin, lipids, or other clinical measures, although this dosing study was not powered to detect such effects. No carryover or adverse effects were observed. CONCLUSIONS Supplementation with seabuckthorn oil augmented in 16:1n-7t and unmodified seabuckthorn oil moderately increased concentrations of their corresponding PLFAs in metabolically healthy adults, supporting the use of supplementation with these fatty acids to test potential clinical effects in humans.This trial was registered at clinicaltrials.gov as NCT02311790.
Collapse
Affiliation(s)
- Neil K Huang
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Nirupa R Matthan
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA,Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA
| | - Jean M Galluccio
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Peilin Shi
- Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA
| | - Alice H Lichtenstein
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA,Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA,Address correspondence to AHL (e-mail: )
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
33
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
34
|
Abstract
Currently, the prevention and treatment of CVD have been a global focus since CVD is the number one cause of mortality and morbidity. In the pathogenesis of CVD, it was generally thought that impaired cholesterol homeostasis might be a risk factor. Cholesterol homeostasis is affected by exogenous factors (i.e. diet) and endogenous factors (i.e. certain receptors, enzymes and transcription factors). In this context, the number of studies investigating the potential mechanisms of dietary fatty acids on cholesterol homeostasis have increased in recent years. As well, the cluster of differentiation 36 (CD36) receptor is a multifunctional membrane receptor involved in fatty acid uptake, lipid metabolism, atherothrombosis and inflammation. CD36 is proposed to be a crucial molecule for cholesterol homeostasis in various mechanisms including absorption/reabsorption, synthesis, and transport of cholesterol and bile acids. Moreover, it has been reported that the amount of fatty acids and fatty acid pattern of the diet influence the CD36 level and CD36-mediated cholesterol metabolism principally in the liver, intestine and macrophages. In these processes, CD36-mediated cholesterol and lipoprotein homeostasis might be impaired by dietary SFA and trans-fatty acids, whereas ameliorated by MUFA in the diet. The effects of PUFA on CD36-mediated cholesterol homeostasis are controversial depending on the amount of n-3 PUFA and n-6 PUFA, and the n-3:n-6 PUFA ratio. Thus, since the CD36 receptor is suggested to be a novel nutrient-sensitive biomarker, the role of CD36 and dietary fatty acids in cholesterol metabolism might be considered in medical nutrition therapy in the near future. Therefore, the novel nutritional target of CD36 and interventions that focus on dietary fatty acids and potential mechanisms underlying cholesterol homeostasis are discussed in this review.
Collapse
|
35
|
Polarization of Macrophages in Human Adipose Tissue is Related to the Fatty Acid Spectrum in Membrane Phospholipids. Nutrients 2019; 12:nu12010008. [PMID: 31861434 PMCID: PMC7020093 DOI: 10.3390/nu12010008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023] Open
Abstract
Residential macrophages in adipose tissue play a pivotal role in the development of inflammation not only within this tissue, but also affect the proinflammatory status of the whole body. Data on human adipose tissue inflammation and the role of macrophages are rather scarce. We previously documented that the proportion of proinflammatory macrophages in human adipose tissue correlates closely with non-HDL cholesterol concentrations. We hypothesized that this is due to the identical influence of diet on both parameters and decided to analyze the fatty acid spectrum in cell membrane phospholipids of the same individuals as a parameter of the diet consumed. Proinflammatory and anti-inflammatory macrophages were isolated from human adipose tissue (n = 43) and determined by flow cytometry as CD14+CD16+CD36high and CD14+CD16−CD163+, respectively. The spectrum of fatty acids in phospholipids in the cell membranes of specimens of the same adipose tissue was analyzed, and the proportion of proinflammatory macrophage increased with the proportions of palmitic and palmitoleic acids. Contrariwise, these macrophages decreased with increasing alpha-linolenic acid, total n-3 fatty acids, n-3/n-6 ratio, and eicosatetraenoic acid. A mirror picture was documented for the proportion of anti-inflammatory macrophages. The dietary score, obtained using a food frequency questionnaire, documented a positive relation to proinflammatory macrophages in individuals who consumed predominantly vegetable fat and fish, and individuals who consumed diets based on animal fat without fish and nut consumption. he present data support our hypothesis that macrophage polarization in human visceral adipose tissue is related to fatty acid metabolism, cell membrane composition, and diet consumed. It is suggested that fatty acid metabolism might participate also in inflammation and the risk of developing cardiovascular disease.
Collapse
|
36
|
Liu X, Zhao K, Yang X, Zhao Y. Gut Microbiota and Metabolome Response of Decaisnea insignis Seed Oil on Metabolism Disorder Induced by Excess Alcohol Consumption. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:10667-10677. [PMID: 31483636 DOI: 10.1021/acs.jafc.9b04792] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This study investigated the modulatory effects of Decaisnea insignis seed oil (DISO), which was rich in palmitoleic acid (55.25%), palmitic acid (12.25%), and oleic acid (28.74%), on alcohol-induced metabolism disorder in mice. Fifty mice were orally administered with 38% alcohol (0.4 mL/day) and without or with DISO (3, 6, and 12 g/kg) for consecutive 12 weeks. DISO inhibited the alcohol-induced weight loss and liver function abnormality (p < 0.01) and shifted the profiles of cecal microbiome: elevating the abundance of Lactobacillus, Ruminoccoceae_UCG_004 (p < 0.05) and decreasing abundance of Parabacteroides (p < 0.05). This treatment also regulated metabolome response of amino acid and lipid metabolism in cecal content: upregulating 5-hydroxyindole-3-acetic acid (p < 0.05), 6-hydroxynicotinic acid, 5-methoxytryptamine, nicotinamide, and nicotinic acid (p < 0.1) and downregulating androsterone, tryptophan, and indole-3-acetamide (p < 0.05). DISO protected against alcoholic liver injury and gut microbiota dysbiosis by enriching the relative abundance of Lactobacillus, which was positively associated with the improvement of intestinal permeability and tryptophan metabolism.
Collapse
|