1
|
Saleh NEH, Ibrahim MY, Saad AH, Abdel-Hakeem EA, Saleh RK, Habeeb WN. The impact of consuming different types of high-caloric fat diet on the metabolic status, liver, and aortic integrity in rats. Sci Rep 2024; 14:18602. [PMID: 39127712 PMCID: PMC11316824 DOI: 10.1038/s41598-024-68299-6] [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: 03/25/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Consumption of high-caloric diets contributes to the alarming number of overweight and obese individuals worldwide, which in turn leads to several diseases and multiple organ dysfunction. Not only has the number of calories taken per day but also the type of fat in the diet has an important impact on health. Accordingly, the purpose of the current study was to examine the impact of different types of high-caloric fat diets on the metabolic status and the integrity of the liver and aorta in albino rats. Adult male albino rats were divided into 6 groups: Control group, long chain-saturated fat group (SFD), long chain-monounsaturated fat (MUFAs) group, long chain-polyunsaturated fat (PUFAs) group, medium-chain fat (MCFAs) group, and short-chain fat (SCFAs) group. Body mass index (BMI), Lee index, and visceral fat amount were reported. Serum levels of insulin, liver transaminases, lipid profile, and different oxidative stress and inflammatory markers were evaluated. Homeostasis Model Assessment of Insulin Resistance (HOMA-IR), and adiponectin/leptin ratio were also calculated. Histopathological examinations of liver and aorta with Masson's trichrome stain, and immune-staining for Nuclear Factor Erythroid-2-Related Factor-2 (Nrf2) were also done. SFD group showed significantly elevated liver transaminases, inflammatory markers, HOMA-IR, dyslipidemia, reduced adiponectin, and deficient anti-oxidative response compared to other groups together with disturbed hepatic and aortic architecture. Other treated groups showed an improvement. PUFAs group showed the highest level of improvement. Not all high-fat diets are hazardous. Diets rich in PUFAs, MUFAs, MCFAs, or SCFAs may protect against the hazards of high caloric diet.
Collapse
Affiliation(s)
| | - Mariam Yahia Ibrahim
- Department of Medical Physiology, Faculty of Medicine, Minia University, El-Minia, 61511, Egypt
| | - Adel Hussein Saad
- Department of Medical Physiology, Faculty of Medicine, Minia University, El-Minia, 61511, Egypt
| | - Elshymaa A Abdel-Hakeem
- Department of Medical Physiology, Faculty of Medicine, Minia University, El-Minia, 61511, Egypt
| | - Rabeh Khairy Saleh
- Department of Pathology, Faculty of Medicine, Minia University, El-Minia, 61511, Egypt
| | - Wagdy N Habeeb
- Department of Medical Physiology, Faculty of Medicine, Minia University, El-Minia, 61511, Egypt
| |
Collapse
|
2
|
Montserrat-de la Paz S, Del Carmen Naranjo M, Lopez S, Del Carmen Millan-Linares M, Rivas-Dominguez A, Jaramillo-Carmona SM, Abia R, Muriana FJG, Bermudez B. Immediate-release niacin and a monounsaturated fatty acid-rich meal on postprandial inflammation and monocyte characteristics in men with metabolic syndrome. Clin Nutr 2023; 42:2138-2150. [PMID: 37774650 DOI: 10.1016/j.clnu.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND & AIM When considered separately, long-term immediate-release niacin and fatty meals enriched in monounsaturated fatty acids (MUFA) decrease postprandial triglycerides, but their effects on postprandial inflammation, which is common in individuals with metabolic syndrome, are less known. Moreover, successful combination is lacking and its impact on acute disorders of the innate immune cells in the metabolic syndrome remains unclear. Here, we aimed to establish the effects from combination with niacin of different fats [butter, enriched in saturated fatty acids (SFA), olive oil, enriched in MUFA, and olive oil supplemented with eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids] on plasma inflammatory markers and circulating monocyte subsets, activation and priming at the postprandial period in individuals with metabolic syndrome. METHODS A random-order within-subject crossover experiment was performed, in which 16 individuals with metabolic syndrome and 16 age-matched healthy volunteers took 2 g immediate-release niacin together with the corresponding fatty meal or a meal with no fat as control. In total, 128 postprandial curves were analysed. We sampled hourly over 6 h for plasma concentrations of soluble inflammatory markers and triglycerides. Circulating monocyte subsets (CD14/CD16 balance), activation (CCL2/CCR2 axis) and priming (M1/M2-like phenotype) at the time of postprandial hypertriglyceridemic peak were also addressed. RESULTS Dietary SFA (combined with niacin) promote postprandial excursions of circulating IL-6, IL-1β, TNF-α and CD14/CCR2-rich monocytes with a pro-inflammatory M1-like phenotype, particularly in individuals with metabolic syndrome. In contrast, dietary MUFA (combined with niacin) postprandially increased circulating CD16-rich monocytes with an anti-inflammatory M2-like phenotype. Omega-3 PUFA did not add to the effects of MUFA. CONCLUSION The co-administration of a single-dose of immediate-release niacin with a fatty meal rich in MUFA, in contrast to SFA, suppresses postprandial inflammation at the levels of both secretory profile and monocyte response in individuals with metabolic syndrome. These findings highlight a potential role of combining niacin and dietary MUFA for the homeostatic control of inflammation and the innate immune system, identifying a new search direction for the management of disorders associated with the metabolic syndrome.
Collapse
Affiliation(s)
- Sergio Montserrat-de la Paz
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain; Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Maria Del Carmen Naranjo
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain; Department of Cell Biology, Faculty of Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS/CSIC), Hospital Universitario Virgen del Rocio, University of Seville, 41013 Seville, Spain
| | - Maria Del Carmen Millan-Linares
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; Cell Biology Unit, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | | | | | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | - Beatriz Bermudez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain; Department of Cell Biology, Faculty of Biology, University of Seville, 41012 Seville, Spain.
| |
Collapse
|
3
|
Machado M, Costa EM, Silva S, Rodriguez-Alcalá LM, Gomes AM, Pintado M. Understanding the Anti-Obesity Potential of an Avocado Oil-Rich Cheese through an In Vitro Co-Culture Intestine Cell Model. Molecules 2023; 28:5923. [PMID: 37570893 PMCID: PMC10421176 DOI: 10.3390/molecules28155923] [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: 06/10/2023] [Revised: 07/27/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
Nowadays, with consumers' requirements shifting towards more natural solutions and the advent of nutraceutical-based approaches, new alternatives for obesity management are being developed. This work aimed to show, for the first time, the potential of avocado oil-fortified cheese as a viable foodstuff for obesity management through complex in vitro cellular models. The results showed that oleic and palmitic acids' permeability through the Caco-2/HT29-MTX membrane peaked at the 2h mark, with the highest apparent permeability being registered for oleic acid (0.14 cm/s). Additionally, the permeated compounds were capable of modulating the metabolism of adipocytes present in the basal compartment, significantly reducing adipokine (leptin) and cytokine (MPC-1, IL-10, and TNF-α) production. The permeates (containing 3.30 µg/mL of palmitic acid and 2.16 µg/mL of oleic acid) also presented an overall anti-inflammatory activity upon Raw 264.7 macrophages, reducing IL-6 and TNF-α secretion. Despite in vivo assays being required, the data showed the potential of a functional dairy product as a valid food matrix to aid in obesity management.
Collapse
Affiliation(s)
| | - Eduardo M. Costa
- CBQF Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | | | | | | | - Manuela Pintado
- CBQF Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| |
Collapse
|
4
|
Farias-Pereira R, Zuk JB, Khavaran H. Plant bioactive compounds from Mediterranean diet improve risk factors for metabolic syndrome. Int J Food Sci Nutr 2023; 74:403-423. [PMID: 37415346 PMCID: PMC10399461 DOI: 10.1080/09637486.2023.2232949] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
Mediterranean (Med) dietary pattern consists of moderate or high consumption of foods that are linked to reduced risk factors for metabolic syndrome (MetS). This comprehensive review evaluates studies on Med diet-representative foods and beverages, such as red wine and olive oil, to understand the inverse associations of Med diet and MetS. The intake of dietary fibre, unsaturated fatty acids, vitamins, and polyphenols - including flavonoids and stilbenes - help to explain the benefits of Med diet on abdominal adiposity, glucose intolerance, hyperlipidaemia, and high blood pressure to some extent. Antioxidant and anti-inflammatory properties of polyphenols as well as the effects of unsaturated fatty acids on lipid metabolism are part of the underlying mechanisms. Overall, this review shows that dietary interventions using Med diet components improve MetS health markers in humans and/or rodents.
Collapse
Affiliation(s)
- Renalison Farias-Pereira
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Joshua B. Zuk
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Hannah Khavaran
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
5
|
Salsinha AS, Socodato R, Relvas JB, Pintado M. The pro- and antiinflammatory activity of fatty acids. BIOACTIVE LIPIDS 2023:51-75. [DOI: 10.1016/b978-0-12-824043-4.00002-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
6
|
Ortega-Gomez A, Lopez S, Varela LM, Jaramillo S, Muriana FJ, Abia R. New evidence for dietary fatty acids in the neutrophil traffic between the bone marrow and the peripheral blood. FOOD CHEMISTRY. MOLECULAR SCIENCES 2022; 5:100133. [PMID: 36111060 PMCID: PMC9467871 DOI: 10.1016/j.fochms.2022.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/11/2022]
Abstract
Chronic administration of a high-fat diet in mice has been established to influence the generation and trafficking of immune cells such as neutrophils in the bone marrow, the dysregulation of which may contribute to a wide range of diseases. However, no studies have tested the hypothesis that a short-term, high-fat diet could early modulate the neutrophil release from bone marrow at fasting and at postprandial in response to a high-fat meal challenge, and that the predominant type of fatty acids in dietary fats could play a role in both context conditions. Based on these premises, we aimed to establish the effects of different fats [butter, enriched in saturated fatty acids (SFAs), olive oil, enriched in monounsaturated fatty acids (MUFAs), and olive oil supplemented with eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids] on neutrophil navigation from bone marrow to blood in mice. The analysis of cellular models for mechanistic understanding and of postprandial blood samples from healthy volunteers for translational purposes was assessed. The results revealed a powerful effect of dietary SFAs in promotion the neutrophil traffic from bone marrow to blood via the CXCL2-CXCR2 axis. Dietary SFAs, but not MUFAs or EPA and DHA, were also associated with increased neutrophil apoptosis and bone marrow inflammation. Similar dietary fatty-acid-induced postprandial neutrophilia was observed in otherwise healthy humans. Therefore, dietary MUFAs might preserve bone marrow health and proper migration of bone marrow neutrophils early in the course of high-fat diets even after the intake of high-fat meals.
Collapse
Key Words
- BMSF, bone marrow supernatant fluid
- Bone marrow inflammation
- Butter
- Ct, threshold cycle
- DHA, docosahexaenoic acid
- Dietary fatty acids
- EPA, eicosapentaenoic acid
- FACS, fluorescence-activated cell sorting
- FSC, forward scatter
- HBSS, Hank’s balance salt solution
- HFDs, high-fat diets
- HSCs, hematopoietic stem cells
- High-fat diets
- LFD, low-fat diet
- MFI, mean fluorescence intensity
- MMP9, matrix metalloproteinase 9
- MUFAs, monounsaturated fatty acids
- Neutrophil mobilisation
- OCM, oral control meal
- OFLs, oral fat loads
- OFMs, oral fat meals
- OSL, oral saline load
- Olive oil
- PI, propidium iodide
- PUFAs, polyunsaturated fatty acids
- SFAs, saturated fatty acids
- SSC, side scatter
- TRLs, triglyceride-rich lipoproteins
- qRT-PCR, quantitative real-time reverse transcription polymerase chain reaction
Collapse
Affiliation(s)
- Almudena Ortega-Gomez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), 41013 Seville, Spain
- Department of Cellular and Molecular Endocrinology, Instituto de Investigacion Biomedica de Malaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Malaga, Spain
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), 41013 Seville, Spain
- Department of Cell Biology, Faculty of Biology, University of Seville, 41012 Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/University of Seville, 41013 Seville, Spain
| | - Lourdes M. Varela
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), 41013 Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/University of Seville, 41013 Seville, Spain
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Sara Jaramillo
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), 41013 Seville, Spain
| | - Francisco J.G. Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), 41013 Seville, Spain
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), 41013 Seville, Spain
| |
Collapse
|
7
|
Li X, Li W, Zhang Z, Wang W, Huang H. SIRT6 overexpression retards renal interstitial fibrosis through targeting HIPK2 in chronic kidney disease. Front Pharmacol 2022; 13:1007168. [PMID: 36172184 PMCID: PMC9510922 DOI: 10.3389/fphar.2022.1007168] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/22/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction: Renal interstitial fibrosis is a common pathophysiological change in the chronic kidney disease (CKD). Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin 6 (SIRT6) is demonstrated to protect against kidney injury. Vitamin B3 is the mostly used form of NAD precursors. However, the role of SIRT6 overexpression in renal interstitial fibrosis of CKD and the association between dietary vitamin B3 intake and renal function remain to be elucidated. Methods: Wild-type (WT) and SIRT6-transgene (SIRT6-Tg) mice were given with high-adenine diets to establish CKD model. HK2 cells were exposed to transforming growth factor β1 (TGF-β1) in vitro to explore related mechanism. Population data from Multi-Ethnic Study of Atherosclerosis (MESA) was used to examine the association between dietary vitamin B3 intake and renal function decline. Results: Compared to WT mice, SIRT6-Tg mice exhibited alleviated renal interstitial fibrosis as evidenced by reduced collagen deposit, collagen I and α-smooth muscle actin expression. Renal function was also improved in SIRT6-Tg mice. Homeodomain interacting protein kinase 2 (HIPK2) was induced during the fibrogenesis in CKD, while HIPK2 was downregulated after SIRT6 overexpression. Further assay in vitro confirmed that SIRT6 depletion exacerbated epithelial-to-mesenchymal transition of HK2 cells, which might be linked with HIPK2 upregulation. HIPK2 was inhibited by SIRT6 in the post-transcriptional level. Population study indicated that higher dietary vitamin B3 intake was independently correlated with a lower risk of estimate glomerular filtration rate decline in those ≥65 years old during follow-up. Conclusion: SIRT6/HIPK2 axis serves as a promising target of renal interstitial fibrosis in CKD. Dietary vitamin B3 intake is beneficial for renal function in the old people.
Collapse
Affiliation(s)
- Xiaoxue Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wenxin Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhengzhipeng Zhang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weidong Wang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Hui Huang,
| |
Collapse
|
8
|
CD4+ and CD8+ T-cell responses in bone marrow to fatty acids in high-fat diets. J Nutr Biochem 2022; 107:109057. [DOI: 10.1016/j.jnutbio.2022.109057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/23/2022] [Accepted: 04/06/2022] [Indexed: 12/30/2022]
|
9
|
Gill PA, Inniss S, Kumagai T, Rahman FZ, Smith AM. The Role of Diet and Gut Microbiota in Regulating Gastrointestinal and Inflammatory Disease. Front Immunol 2022; 13:866059. [PMID: 35450067 PMCID: PMC9016115 DOI: 10.3389/fimmu.2022.866059] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022] Open
Abstract
Diet is an important lifestyle factor that is known to contribute in the development of human disease. It is well established that poor diet plays an active role in exacerbating metabolic diseases, such as obesity, diabetes and hypertension. Our understanding of how the immune system drives chronic inflammation and disease pathogenesis has evolved in recent years. However, the contribution of dietary factors to inflammatory conditions such as inflammatory bowel disease, multiple sclerosis and arthritis remain poorly defined. A western diet has been associated as pro-inflammatory, in contrast to traditional dietary patterns that are associated as being anti-inflammatory. This may be due to direct effects of nutrients on immune cell function. Diet may also affect the composition and function of gut microbiota, which consequently affects immunity. In animal models of inflammatory disease, diet may modulate inflammation in the gastrointestinal tract and in other peripheral sites. Despite limitations of animal models, there is now emerging evidence to show that anti-inflammatory effects of diet may translate to human gastrointestinal and inflammatory diseases. However, appropriately designed, larger clinical studies must be conducted to confirm the therapeutic benefit of dietary therapy.
Collapse
Affiliation(s)
- Paul A Gill
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| | - Saskia Inniss
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| | - Tomoko Kumagai
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| | - Farooq Z Rahman
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom.,Department of Gastroenterology, University College London Hospitals National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Andrew M Smith
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| |
Collapse
|
10
|
Pan B, Liu X, Shi J, Chen Y, Xu Z, Shi D, Ruan G, Wang F, Huang Y, Xu C. A Meta-Analysis of Microbial Therapy Against Metabolic Syndrome: Evidence From Randomized Controlled Trials. Front Nutr 2021; 8:775216. [PMID: 34977119 PMCID: PMC8714845 DOI: 10.3389/fnut.2021.775216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
Background and aims: Metabolic syndrome (MetS), accompanied with significant intestinal dysbiosis, causes a great public health burden to human society. Here, we carried out a meta-analysis to qualify randomized controlled trials (RCTs) and to systematically evaluate the effect of microbial therapy on MetS. Methods and results: Forty-two RCTs were eligible for this meta-analysis after searching the PubMed, Cochrane, and Embase databases. Pooled estimates demonstrated that treatment with microbial therapy significantly reduced the waist circumference (WC) (SMD = -0.26, 95% CI -0.49, -0.03), fasting blood glucose (FBG) (SMD = -0.35, 95% CI -0.52, -0.18), total cholesterol (TC) (SMD = -0.36, 95% CI -0.55, -0.17), low-density lipoprotein cholesterol (LDL-C) (SMD = -0.42, 95% CI -0.61, -0.22), and triacylglycerol (TG)(SMD = -0.38, 95% CI -0.55, -0.20), but increased the high-density lipoprotein cholesterol (HDL-C) (SMD = 0.28, 95% CI.03, 0.52). Sensitivity analysis indicated that after eliminating one study utilizing Bifidobacteriumlactis, results became statistically significant in diastolic blood pressure (DBP) (SMD = -0.24, 95% CI -0.41, -0.07) and in Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) (SMD = -0.28, 95% CI -0.54, -0.03), while the body mass index (BMI) showed significant difference after eliminating one study utilizing oat bran (SMD = -0.16, 95% CI -0.31, -0.01). There was still no significant effect in systolic blood pressure (SBP) and in hemoglobin A1c (HbA1c%). Conclusion: In patients with MetS, the conditioning with microbial therapy notably improves FBG, TC, TG, HDL-C, LDL-C, WC, BMI (except for the study using oat bran), HOMA-IR, and DBP (except for the Study using Bifidobacteriumlactis), however, with no effect in SBP and in HbA1c%.
Collapse
Affiliation(s)
- Binhui Pan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiujie Liu
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Jiangmin Shi
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaoxuan Chen
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhihua Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dibang Shi
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaoyi Ruan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, China
| | - Yingpeng Huang
- Department of Gastrointestinal Oncology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Changlong Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Lemus-Conejo A, Medrano M, Lopez S, Millan-Linares MC, Rosillo MA, Perez-Simon JA, Muriana FJG, Abia R. MUFAs in High-Fat Diets Protect against Obesity-Induced Bias of Hematopoietic Cell Lineages. Mol Nutr Food Res 2021; 65:e2001203. [PMID: 34132459 DOI: 10.1002/mnfr.202001203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/27/2021] [Indexed: 11/08/2022]
Abstract
SCOPE The role of dietary fatty acids in the generation of bone marrow (BM) immune cells and their trafficking to extramedullary compartments in the obesity is not yet fully understood. METHODS AND RESULTS C57BL/6J mice are randomly assigned to isocaloric high-fat diets (HFDs) formulate with dietary fats rich in saturated fatty acids (SFAs), monounsaturated fatty acids (MUFAs) or MUFAs fortified with eicosapentaenoic and docosahexaenoic acids for 20 weeks, followed by profiling of the obese metabolic phenotype and immunophenotypic features of immune cells in blood, spleen, and BM. All HFDs induce an obese phenotype, but it becomes largely less disruptive after the HFDs are enriched in MUFAs, which also induce signs of granulopoiesis and an expansion of long-term hematopoietic stem and granulocyte-macrophage progenitor cells in BM. In contrast, a HFD enriched in SFAs disturbs the fitness of medullary lymphocytes and promotes monopoiesis in favor of pro-inflammatory activated subsets. CONCLUSION The reshaping of the fatty acid pools with MUFAs from the diet serves to manipulate the generation and trafficking of immune cells that are biased during obesity. These findings reveal a novel strategy by which dietary MUFAs may be instrumental in combating HFD-induced dysfunctional immune systems.
Collapse
Affiliation(s)
- Ana Lemus-Conejo
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| | - Mayte Medrano
- Department of Haematology, Instituto de Biomedicina de Sevilla (IBiS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocio, University of Seville, Seville, 41012, Spain
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
- Department of Cell Biology, Faculty of Biology, University of Seville, Seville, 41012, Spain
- Instituto de Biomedicina de Sevilla (IBiS/CSIC), Hospital Universitario Virgen del Rocio, University of Seville, Seville, 41012, Spain
| | | | - Maria A Rosillo
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| | - Jose A Perez-Simon
- Department of Haematology, Instituto de Biomedicina de Sevilla (IBiS/CSIC/CIBERONC), Hospital Universitario Virgen del Rocio, University of Seville, Seville, 41012, Spain
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, The Spanish National Research Council (CSIC), Seville, 41013, Spain
| |
Collapse
|
12
|
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: 115] [Impact Index Per Article: 23.0] [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
|
13
|
Vazquez-Madrigal C, Lopez S, Grao-Cruces E, Millan-Linares MC, Rodriguez-Martin NM, Martin ME, Alba G, Santa-Maria C, Bermudez B, Montserrat-de la Paz S. Dietary Fatty Acids in Postprandial Triglyceride-Rich Lipoproteins Modulate Human Monocyte-Derived Dendritic Cell Maturation and Activation. Nutrients 2020; 12:nu12103139. [PMID: 33066622 PMCID: PMC7656296 DOI: 10.3390/nu12103139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Dietary fatty acids have been demonstrated to modulate systemic inflammation and induce the postprandial inflammatory response of circulating immune cells. We hypothesized that postprandial triglyceride-rich lipoproteins (TRLs) may have acute effects on immunometabolic homeostasis by modulating dendritic cells (DCs), sentinels of the immunity that link innate and adaptive immune systems. In healthy volunteers, saturated fatty acid (SFA)-enriched meal raised serum levels of granulocyte/macrophage colony-stimulating factor GM-CSF (SFAs > monounsaturated fatty acids (MUFAs) = polyunsaturated fatty acids (PUFAs)) in the postprandial period. Autologous TRL-SFAs upregulated the gene expression of DC maturation (CD123 and CCR7) and DC pro-inflammatory activation (CD80 and CD86) genes while downregulating tolerogenic genes (PD-L1 and PD-L2) in human monocyte-derived DCs (moDCs). These effects were reversed with oleic acid-enriched TRLs. Moreover, postprandial SFAs raised IL-12p70 levels, while TRL-MUFAs and TRL-PUFAs increased IL-10 levels in serum of healthy volunteers and in the medium of TRL-treated moDCs. In conclusion, postprandial TRLs are metabolic entities with DC-related tolerogenic activity, and this function is linked to the type of dietary fat in the meal. This study shows that the intake of meals enriched in MUFAs from olive oil, when compared with meals enriched in SFAs, prevents the postprandial production and priming of circulating pro-inflammatory DCs, and promotes tolerogenic response in healthy subjects. However, functional assays with moDCs generated in the presence of different fatty acids and T cells could increase the knowledge of postprandial TRLs’ effects on DC differentiation and function.
Collapse
Affiliation(s)
- Carlos Vazquez-Madrigal
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Soledad Lopez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Elena Grao-Cruces
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Maria C. Millan-Linares
- Department of Food & Health, Instituto de la Grasa, CSIC, 41013 Seville, Spain; (M.C.M.-L.); (N.M.R.-M.)
| | - Noelia M. Rodriguez-Martin
- Department of Food & Health, Instituto de la Grasa, CSIC, 41013 Seville, Spain; (M.C.M.-L.); (N.M.R.-M.)
| | - Maria E. Martin
- Department of Cell Biology, Faculty of Biology, Universidad de Sevilla, 41012 Seville, Spain; (M.E.M.); (B.B.)
| | - Gonzalo Alba
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Consuelo Santa-Maria
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain;
| | - Beatriz Bermudez
- Department of Cell Biology, Faculty of Biology, Universidad de Sevilla, 41012 Seville, Spain; (M.E.M.); (B.B.)
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
- Correspondence: ; Tel.: +34-954-559-850
| |
Collapse
|
14
|
Mitigating Effect of 1-Palmitoyl-2-Linoleoyl-3-Acetyl-Rac-Glycerol (PLAG) on a Murine Model of 5-Fluorouracil-Induced Hematological Toxicity. Cancers (Basel) 2019; 11:cancers11111811. [PMID: 31752148 PMCID: PMC6896120 DOI: 10.3390/cancers11111811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 12/19/2022] Open
Abstract
5-Fluorouracil (5-FU) is an antimetabolite chemotherapy widely used for the treatment of various cancers. However, many cancer patients experience hematological side effects following 5-FU treatment. Here, we investigated the protective effects of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) as a mitigator against 5-FU-induced hematologic toxicity, including neutropenia, monocytopenia, thrombocytopenia, and thrombocytosis, in Balb/c mice injected with 5-FU (100 mg/kg, i.p.). Administration of PLAG significantly and dose-dependently reduced the duration of neutropenia and improved the nadirs of absolute neutrophil counts (ANCs). Moreover, while the ANCs of all mice in the control fell to the severely neutropenic range, none of the mice in the PLAG 200 and 400 mg/kg-treated groups experienced severe neutropenia. Administration of PLAG significantly delayed the mean first day of monocytopenia and reduced the duration of monocytopenia. PLAG also effectively reduced extreme changes in platelet counts induced by 5-FU treatment, thus preventing 5-FU-induced thrombocytopenia and thrombocytosis. PLAG significantly decreased plasma levels of the chemokine (C–X–C motif) ligand 1 (CXCL1), CXCL2, interleukin (IL)-6, and C-reactive protein (CRP), which were elevated consistently with the occurrence time of neutropenia, monocytopenia, and thrombocytopenia. When compared with olive oil and palmitic linoleic hydroxyl glycerol (PLH), only PLAG effectively mitigated 5-FU-induced hematological toxicity, indicating that it has a distinctive mechanism of action. In conclusion, PLAG may have therapeutic potential as a mitigator for 5-FU-induced neutropenia and other hematological disorders.
Collapse
|