151
|
Ciumărnean L, Milaciu MV, Runcan O, Vesa ȘC, Răchișan AL, Negrean V, Perné MG, Donca VI, Alexescu TG, Para I, Dogaru G. The Effects of Flavonoids in Cardiovascular Diseases. Molecules 2020; 25:E4320. [PMID: 32967119 PMCID: PMC7571023 DOI: 10.3390/molecules25184320] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/13/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Flavonoids are metabolites of plants and fungus. Flavonoid research has been paid special attention to in recent times after the observation of their beneficial effects on the cardiovascular system. These favorable effects are exerted by flavonoids mainly due to their antioxidant properties, which result from the ability to decrease the oxidation of low-density lipoproteins, thus improving the lipid profiles. The other positive effect exerted on the cardiovascular system is the ability of flavonoids to produce vasodilation and regulate the apoptotic processes in the endothelium. Researchers suggested that these effects, including their anti-inflammatory function, are consequences of flavonoids' potent antioxidant properties, but recent studies have shown multiple signaling pathways linked to them, thus suggesting that there are more mechanisms involved in the beneficial effect of the flavonoids on the human body. This review aims to present the latest data on the classification of these substances, their main mechanisms of action in the human body, and the beneficial effects on the physiological and pathological status of the cardiovascular system.
Collapse
Affiliation(s)
- Lorena Ciumărnean
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (M.-G.P.); (V.N.); (T.-G.A.); (I.P.)
| | - Mircea Vasile Milaciu
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (M.-G.P.); (V.N.); (T.-G.A.); (I.P.)
| | - Octavia Runcan
- Regional Institute of Gastroenterology and Hepatology ‘Octavian Fodor’ Cluj-Napoca, 400162 Cluj-Napoca, Romania;
| | - Ștefan Cristian Vesa
- Department 2—Functional Sciences, Discipline of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andreea Liana Răchișan
- Department of Pediatrics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400177 Cluj-Napoca, Romania
| | - Vasile Negrean
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (M.-G.P.); (V.N.); (T.-G.A.); (I.P.)
| | - Mirela-Georgiana Perné
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (M.-G.P.); (V.N.); (T.-G.A.); (I.P.)
| | - Valer Ioan Donca
- Department of Geriatrics-Gerontology, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania;
| | - Teodora-Gabriela Alexescu
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (M.-G.P.); (V.N.); (T.-G.A.); (I.P.)
| | - Ioana Para
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (L.C.); (M.V.M.); (M.-G.P.); (V.N.); (T.-G.A.); (I.P.)
| | - Gabriela Dogaru
- Department of Physical Medicine & Rehabilitation, “Iuliu Hațieganu” University of Medicine and Pharmacy, Louis Pasteur Street 6, 400349 Cluj-Napoca, Romania;
| |
Collapse
|
152
|
Huang W, Wu X, Xue Y, Zhou Y, Xiang H, Yang W, Wei Y. MicroRNA-3614 regulates inflammatory response via targeting TRAF6-mediated MAPKs and NF-κB signaling in the epicardial adipose tissue with coronary artery disease. Int J Cardiol 2020; 324:152-164. [PMID: 32950591 DOI: 10.1016/j.ijcard.2020.09.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The inflammatory status of epicardial adipose tissue (EAT) is one of the factors leading to the development of related diseases such as coronary artery disease (CAD). The thickness of CAD EAT increases and is accompanied with increased macrophage infiltration and heightened inflammatory responses. However, microRNAs (miRNAs) regulating the inflammatory responses of macrophages in CAD EAT remain unclear. METHOD miRNA expression profiles of CAD EATs and non-CAD EATs were determined by miRNA microarrays. Quantitative real-time reverse transcription-polymerase chain reaction, Western blotting, immunohistochemical assay, and fluorescence in-situ hybridization were adopted to detect miR-3614 expression and function in EATs and macrophages. The interaction between miR-3614 and tumor necrosis factor receptor-associated factor 6 (TRAF6) was identified using an online website combined with a dual-luciferase reporter assay. Enzyme-linked immunosorbent assay was performed to detect the expression of inflammatory cytokines. RESULTS The decreased expression of miR-3614 was identified in CAD EAT. The level of miR-3614 was down-regulated by lipopolysaccharide (LPS) in macrophages, whereas LPS-induced inflammatory injury can be reduced by miR-3614 overexpression. TRAF6 was predicted and verified to be a target of miR-3614. The phosphorylated levels of kinases in the mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB pathways were inhibited by miR-3614 overexpression. Importantly, the knockdown of TRAF6 inhibited the LPS-induced inflammatory cytokine expressions in cells. CONCLUSION A novel negative feedback loop by miR-3614 possibly contribute to the regulation of inflammatory processes via targeting the TRAF6/MAPK/NF-κB pathway in EATs and prevents an overwhelming inflammatory response.
Collapse
Affiliation(s)
- Wenhua Huang
- Department of Thoracic and Cardiovascular Surgery, Ganzhou Municipal Hospital, Ganzhou, Jiangxi 341000, China
| | - Xinggang Wu
- Medicine Department, Shihezi University, Shihezi, Xinjiang 832000, China
| | - Yajun Xue
- Medicine Department, Shihezi University, Shihezi, Xinjiang 832000, China; Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, Xinjiang 832000, China
| | - Yijun Zhou
- Medicine Department, Shihezi University, Shihezi, Xinjiang 832000, China
| | - Hui Xiang
- Medicine Department, Shihezi University, Shihezi, Xinjiang 832000, China
| | - Wenkai Yang
- Department of Cardiovascular Surgery, Affiliated Central People's Hospital of Zhanjiang, Guangdong Medical University, Zhanjiang 524045, China
| | - Yutao Wei
- Department of Thoracic Surgery, Jining First People's Hospital, Jining, Shandong 250000, China.
| |
Collapse
|
153
|
Perazza LR, Daniel N, Dubois MJ, Pilon G, Varin TV, Blais M, Martinez Gonzales JL, Bouchard M, Asselin C, Lessard M, Pouliot Y, Roy D, Marette A. Distinct Effects of Milk-Derived and Fermented Dairy Protein on Gut Microbiota and Cardiometabolic Markers in Diet-Induced Obese Mice. J Nutr 2020; 150:2673-2686. [PMID: 32886125 PMCID: PMC7549311 DOI: 10.1093/jn/nxaa217] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/17/2020] [Accepted: 07/01/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Recent meta-analyses suggest that the consumption of fermented dairy products reduces type 2 diabetes and cardiovascular disease (CVD) risk, although the underlying mechanisms remain unclear. OBJECTIVE We evaluated whether dairy protein products modulated gut microbiota and cardiometabolic features in mouse models of diet-induced obesity and CVD. METHODS Eight-week-old C57BL/6J wild-type (WT) and LDLr-/-ApoB100/100 (LRKO) male mice were fed for 12 and 24 wk, respectively, with a high-fat/high-sucrose diet [66% kcal lipids, 22% kcal carbohydrates (100% sucrose), 12% kcal proteins]. The protein sources of the 4 diets were 100% nondairy protein (NDP), or 50% of the NDP energy replaced by milk (MP), milk fermented by Lactobacillus helveticus (FMP), or Greek-style yogurt (YP) protein. Fecal 16S rRNA gene-based amplicon sequencing, intestinal gene expression, and glucose tolerance test were conducted. Hepatic inflammation and circulating adhesion molecules were measured by multiplex assays. RESULTS Feeding WT mice for 12 wk led to a 74% increase in body weight, whereas after 24 wk the LRKO mice had a 101.5% increase compared with initial body weight. Compared with NDP and MP, the consumption of FMP and YP modulated the gut microbiota composition in a similar clustering pattern, upregulating the Streptococcus genus in both genotypes. In WT mice, feeding YP compared with NDP increased the expression of genes involved in jejunal (Reg3b, 7.3-fold, P = 0.049) and ileal (Ocln, 1.7-fold, P = 0.047; Il1-β,1.7-fold, P = 0.038; Nos2, 3.8-fold, P = 0.018) immunity and integrity. In LRKO mice, feeding YP compared with MP improved insulin sensitivity by 65% (P = 0.039). In LRKO mice, feeding with FMP versus NDP attenuated hepatic inflammation (monocyte chemoattractant protein 1, 2.1-fold, P ˂ 0.0001; IL1-β, 5.7-fold, P = 0.0003; INF-γ, 1.7-fold, P = 0.002) whereas both FMP [vascular adhesion molecule 1 (VCAM1), 1.3-fold, P = 0.0003] and YP (VCAM1, 1.04-fold, P = 0.013; intracellular adhesion molecule 1, 1.4-fold, P = 0.028) decreased circulating adhesion molecules. CONCLUSION Both fermented dairy protein products reduce cardiometabolic risk factors in diet-induced obese mice, possibly by modulating the gut microbiota.
Collapse
Affiliation(s)
- Laís Rossi Perazza
- Faculty of Medicine, Laval University, Quebec City, Quebec, Canada,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Noëmie Daniel
- Faculty of Medicine, Laval University, Quebec City, Quebec, Canada,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Marie-Julie Dubois
- Faculty of Medicine, Laval University, Quebec City, Quebec, Canada,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Geneviève Pilon
- Faculty of Medicine, Laval University, Quebec City, Quebec, Canada,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Thibault Vincent Varin
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Mylène Blais
- Sherbrooke R & D Center, Agriculture and Agri-Food Canada, Sherbrooke, Quebec, Canada
| | | | - Michaël Bouchard
- Sherbrooke R & D Center, Agriculture and Agri-Food Canada, Sherbrooke, Quebec, Canada
| | - Claude Asselin
- Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Quebec, Canada
| | - Martin Lessard
- Sherbrooke R & D Center, Agriculture and Agri-Food Canada, Sherbrooke, Quebec, Canada
| | - Yves Pouliot
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Denis Roy
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | | |
Collapse
|
154
|
Fleischhacker SE, Woteki CE, Coates PM, Hubbard VS, Flaherty GE, Glickman DR, Harkin TR, Kessler D, Li WW, Loscalzo J, Parekh A, Rowe S, Stover PJ, Tagtow A, Yun AJ, Mozaffarian D. Strengthening national nutrition research: rationale and options for a new coordinated federal research effort and authority. Am J Clin Nutr 2020; 112:721-769. [PMID: 32687145 PMCID: PMC7454258 DOI: 10.1093/ajcn/nqaa179] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The US faces remarkable food and nutrition challenges. A new federal effort to strengthen and coordinate nutrition research could rapidly generate the evidence base needed to address these multiple national challenges. However, the relevant characteristics of such an effort have been uncertain. OBJECTIVES Our aim was to provide an objective, informative summary of 1) the mounting diet-related health burdens facing our nation and corresponding economic, health equity, national security, and sustainability implications; 2) the current federal nutrition research landscape and existing mechanisms for its coordination; 3) the opportunities for and potential impact of new fundamental, clinical, public health, food and agricultural, and translational scientific discoveries; and 4) the various options for further strengthening and coordinating federal nutrition research, including corresponding advantages, disadvantages, and potential executive and legislative considerations. METHODS We reviewed government and other published documents on federal nutrition research; held various discussions with expert groups, advocacy organizations, and scientific societies; and held in-person or phone meetings with >50 federal staff in executive and legislative roles, as well as with a variety of other stakeholders in academic, industry, and nongovernment organizations. RESULTS Stark national nutrition challenges were identified. More Americans are sick than are healthy, largely from rising diet-related illnesses. These conditions create tremendous strains on productivity, health care costs, health disparities, government budgets, US economic competitiveness, and military readiness. The coronavirus disease 2019 (COVID-19) outbreak has further laid bare these strains, including food insecurity, major diet-related comorbidities for poor outcomes from COVID-19 such as diabetes, hypertension, and obesity, and insufficient surveillance on and coordination of our food system. More than 10 federal departments and agencies currently invest in critical nutrition research, yet with relatively flat investments over several decades. Coordination also remains suboptimal, documented by multiple governmental reports over 50 years. Greater harmonization and expansion of federal investment in nutrition science, not a silo-ing or rearrangement of existing investments, has tremendous potential to generate new discoveries to improve and sustain the health of all Americans. Two identified key strategies to achieve this were as follows: 1) a new authority for robust cross-governmental coordination of nutrition research and other nutrition-related policy and 2) strengthened authority, investment, and coordination for nutrition research within the NIH. These strategies were found to be complementary, together catalyzing important new science, partnerships, coordination, and returns on investment. Additional complementary actions to accelerate federal nutrition research were identified at the USDA. CONCLUSIONS The need and opportunities for strengthened federal nutrition research are clear, with specific identified options to help create the new leadership, strategic planning, coordination, and investment the nation requires to address the multiple nutrition-related challenges and grasp the opportunities before us.
Collapse
Affiliation(s)
| | - Catherine E Woteki
- University of Virginia Biocomplexity Institute and Initiative, Arlington, VA, USA
| | - Paul M Coates
- Retired, National Institutes of Health, Bethesda, MD, USA
| | - Van S Hubbard
- Retired, National Institutes of Health, Bethesda, MD, USA
| | - Grace E Flaherty
- Gerald J and Dorothy R Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA
| | | | | | - David Kessler
- Former Food and Drug Administration Commissioner, College Park, MD, USA
| | | | - Joseph Loscalzo
- Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Patrick J Stover
- Texas A&M AgriLife, Texas A&M College of Agriculture and Life Sciences, and Texas A&M AgriLife Research, College Station, TX, USA
| | | | | | - Dariush Mozaffarian
- Gerald J and Dorothy R Friedman School of Nutrition Science and Policy at Tufts University, Boston, MA, USA
| |
Collapse
|
155
|
Sharif H, Akash MSH, Rehman K, Irshad K, Imran I. Pathophysiology of atherosclerosis: Association of risk factors and treatment strategies using plant-based bioactive compounds. J Food Biochem 2020; 44:e13449. [PMID: 32851658 DOI: 10.1111/jfbc.13449] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Under physiological conditions, endothelial cells act as protective barrier which prevents direct contact of blood with circulating factors via production of tissue plasminogen activator. Risk factors of metabolic disorders are responsible to induce endothelial dysfunction and may consequently lead to prognosis of atherosclerosis. This article summarizes the process of atherosclerosis which involves number of sequences including formation and interaction of AGE-RAGE, activation of polyol pathway, protein kinase C, and hexosamine-mediated pathway. All these mechanisms can lead to the development of oxidative stress which may further aggravate condition. Different pharmacological interventions are being used to treat atherosclerosis, however, these might be associated with mild to severe side effects. Therefore, plant-based bioactive compounds having potential to combat and prevent atherosclerosis in diabetic patients are attaining recent focus. By understanding process of development and mechanisms involved in atherosclerotic plaque formation, these bioactive compounds can be better option for future therapeutic interventions for atherosclerosis treatment. PRACTICAL APPLICATIONS: Atherosclerosis is one of major underlying disorders of cardiovascular diseases which occur through multiple mechanisms and is associated with metabolic disorders. Conventional therapeutic interventions are not only used to treat atherosclerosis, but are also commonly associated with mild to severe side effects. Therefore, nowadays, bioactive compounds having potential to combat and prevent atherosclerosis in diabetic patients are preferred. By understanding mechanisms involved in atherosclerotic plaque formation, bioactive compounds can be better understood for treatment of atherosclerosis. In this manuscript, we have focused on treatment strategies of atherosclerosis using bioactive compounds notably alkaloids and flavonoids having diverse pharmacological and therapeutic potentials with special focus on the mechanism of action of these bioactive compounds suitable for treatment of atherosclerosis. This manuscript will provide the scientific insights of bioactive compounds to researchers who are working in the area of drug discovery and development to control pathogenesis and development of atherosclerosis and its associated cardiometabolic disorders.
Collapse
Affiliation(s)
- Hina Sharif
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Kanwal Irshad
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Imran Imran
- Department of Pharmacology, Bahauddin Zakariya University, Multan, Pakistan
| |
Collapse
|
156
|
Ghosh S, He W, Gao J, Luo D, Wang J, Chen J, Huang H. Whole milk consumption is associated with lower risk of coronary artery calcification progression: evidences from the Multi-Ethnic Study of Atherosclerosis. Eur J Nutr 2020; 60:1049-1058. [PMID: 32583016 DOI: 10.1007/s00394-020-02301-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Coronary artery calcification (CAC) progression is a strong predictor of cardiovascular disease (CVD) morbidity and mortality. However, the association between whole milk and CAC progression remains unknown. Recent studies highlighted beneficial effects of short chain fatty acids (SCFA) from whole milk on CVD. In this study, we attempted to investigate the relationship between whole milk consumption and CAC progression, and the potential effect of SCFA in it. METHODS We analyzed a population-based cohort with 5273 participants from the Multi-Ethnic Study of Atherosclerosis (MESA) who completed a dietary questionnaire at baseline. CAC was measured at baseline and subsequent follow-up examinations by multi-detector computed tomography (MDCT) scans with Agatston scores. CAC progression was defined as increased CAC scores in the follow-up from the baseline exam. RESULTS Participants consuming whole milk exhibited lower baseline CAC and CAC progression than those who never/rarely consumed whole milk (P < 0.001 and P = 0.010, respectively). Moreover, multivariable logistic regression analysis demonstrated that whole milk intake was independently associated with lower CAC progression (OR 0.765; 95% CI 0.600-0.977; P = 0.032), especially in males, participants with age ≤ 64 years and with body mass index (BMI) ≤ 25 kg/m2. Mediation analysis further showed that caproic acid, one kind of SCFA, partly mediated protective effects of whole milk on CAC progression. CONCLUSIONS Self-reported whole milk consumption was inversely associated with CAC progression in community-dwelling participants, especially in those at relatively low cardiovascular risks. The beneficial effect was partially mediated by SCFA. Therefore, whole milk can be incorporated into part of a cardio-protective diet. Regarding this, future studies may target SCFA to provide insight into more mechanistic views.
Collapse
Affiliation(s)
- Sounak Ghosh
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Wanbing He
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jingwei Gao
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dongling Luo
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jingfeng Wang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Chen
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Hui Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
157
|
Zhang L, Wang P, Shi M, Fang Z, Ji J, Liao X, Hu X, Chen F. The modulation of Luffa cylindrica (L.) Roem supplementation on gene expression and amino acid profiles in liver for alleviating hepatic steatosis via gut microbiota in high-fat diet-fed mice: insight from hepatic transcriptome analysis. J Nutr Biochem 2020; 80:108365. [DOI: 10.1016/j.jnutbio.2020.108365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/03/2020] [Accepted: 02/21/2020] [Indexed: 12/13/2022]
|
158
|
Nwabuo CC, Betoko AS, Reis JP, Moreira HT, Vasconcellos HD, Guallar E, Cox C, Sidney S, Ambale-Venkatesh B, Lewis CE, Schreiner PJ, Lloyd-Jones D, Kiefe CI, Gidding SS, Lima JAC. Coffee and tea consumption in the early adult lifespan and left ventricular function in middle age: the CARDIA study. ESC Heart Fail 2020; 7:1510-1519. [PMID: 32449612 PMCID: PMC7373924 DOI: 10.1002/ehf2.12684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/01/2020] [Accepted: 03/08/2020] [Indexed: 01/12/2023] Open
Abstract
Aims The long‐term impact of coffee or tea consumption on subclinical left ventricular (LV) systolic or diastolic function has not been previously studied. We examined the association between coffee or tea consumption beginning in early adulthood and cardiac function in midlife. Methods and results We investigated 2735 Coronary Artery Risk Development in Young Adults (CARDIA) study participants with long‐term total caffeine intake, coffee, and tea consumption data from three visits over a 20 year interval and available echocardiography indices at the CARDIA Year‐25 exam (2010–2011). Linear regression models were used to assess the association between caffeine intake, tea, and coffee consumption (independent variables) and echocardiography outcomes [LV mass, left atrial volume, and global longitudinal strain (GLS), LV ejection fraction (LVEF), and transmitral Doppler early filling velocity to tissue Doppler early diastolic mitral annular velocity (E/e´)]. Models were adjusted for standard cardiovascular risk factors, socioeconomic status, physical activity, alcohol use, and dietary factors (calorie intake, whole and refined grain intake, and fruit and vegetable consumption). Mean (standard deviation) age was 25.2 (3.5) years at the CARDIA Year‐0 exam (1985–1986), 57.4% were women, and 41.9% were African‐American. In adjusted multivariable linear regression models assessing the relationship between coffee consumption and GLS, beta coefficients when comparing coffee drinkers of <1, 1–2, 3–4, and >4 cups/day with non‐coffee drinkers were β = −0.30%, P < 0.05; β = −0.35%, P < 0.05; β = −0.32%, P < 0.05; β = −0.40%, P > 0.05; respectively (more negative values implies better systolic function). In adjusted multivariable linear regression models assessing the relationship between coffee consumption and E/e´, beta coefficients when comparing coffee drinkers of <1, 1–2, 3–4, and >4 cups/day with non‐coffee drinkers were β = −0.29, P < 0.05; β = −0.38, P < 0.01; β = −0.20, P > .05; and β = −0.37, P > 0.05, respectively (more negative values implies better diastolic function). High daily coffee consumption (>4 cups/day) was associated with worse LVEF (β = −1.69, P < 0.05). There were no associations between either tea drinking or total caffeine intake and cardiac function (P > 0.05 for all). Conclusions Low‐to‐moderate daily coffee consumption from early adulthood to middle age was associated with better LV systolic and diastolic function in midlife. High daily coffee consumption (>4cups/day) was associated with worse LV function. There was no association between caffeine or tea intake and cardiac function.
Collapse
Affiliation(s)
- Chike C Nwabuo
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Aisha S Betoko
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jared P Reis
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henrique T Moreira
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Ribeirão Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Henrique D Vasconcellos
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Eliseo Guallar
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher Cox
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen Sidney
- Division of Research, Kaiser Permanente, Oakland, CA, USA
| | | | - Cora E Lewis
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Catarina I Kiefe
- Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, USA
| | - Samuel S Gidding
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - João A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
159
|
Wang XY, Liu FC, Yang XL, Li JX, Cao J, Lu XF, Huang JF, Li Y, Chen JC, Zhao LC, Shen C, Hu DS, Zhao YX, Yu L, Liu XQ, Wu XP, Gu DF. Association of cardiovascular diseases with milk intake among general Chinese adults. Chin Med J (Engl) 2020; 133:1144-1154. [PMID: 32433046 PMCID: PMC7249710 DOI: 10.1097/cm9.0000000000000786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The association of milk intake with cardiovascular disease (CVD) and cause-specific mortality remained controversial and evidence among the Chinese population was limited. We aimed to study the relationship between milk intake and CVDs among general Chinese adults. METHODS A total of 104,957 participants received questionnaire survey. Results of physical examination such as anthropometric measurements and biochemical tests during 2007 to 2008, demographic data and their information on milk intake were collected through standardized questionnaires. Cox proportional hazard regression models were used to calculate hazard ratios (HRs) and their corresponding 95% confidence intervals (CIs) of CVD incidence, cause-specific mortality and all-cause mortality related to milk intake. Restricted cubic splines (RCSs) were applied to examine dose-response associations. RESULTS Among the 91,757 participants with a median follow-up period of 5.8 years, we documented 3877 CVD cases and 4091 all-cause deaths. Compared with participants who never consumed milk, the multivariate-adjusted HRs (95% CIs) of CVD incidence for 1 to 150 g/day, 151 to 299 g/day, and ≥300 g/day were 0.94 (0.86-1.03) (P > 0.05), 0.77 (0.66-0.89) (P < 0.05), and 0.59 (0.40-0.89) (P < 0.05), respectively; each 100 g increase of daily milk intake was associated with 11% lower risk of CVD incidence (HR, 0.89; 95% CI: 0.85-0.94; P < 0.001), and 11% lower risk of CVD mortality (HR, 0.89; 95% CI: 0.82-0.97; P = 0.008) after adjustment for age, sex, residential area, geographic region, education level, family history of CVD, smoking, alcohol drinking, physical activity level, body mass index, and healthy diet status (ideal or not). RCS analyses also showed a linear dose-response relationship with CVD (P for overall significance of the curve <0.001; P for non-linearity = 0.979; P for linearity <0.001) and stroke (P for overall significance of the curve = 0.010; P for non-linearity = 0.998; P for linearity = 0.002) incidence, and CVD mortality (P for overall significance of the curve = 0.045; P for non-linearity = 0.768; P for linearity = 0.014) within the current range of daily milk intake. CONCLUSIONS Daily milk intake was associated with lower risk of CVD incidence and mortality in a linear inverse relationship. The findings provide new evidence for dietary recommendations in CVD prevention among Chinese adults and people with similar dietary pattern in other countries.
Collapse
Affiliation(s)
- Xin-Yan Wang
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Fang-Chao Liu
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xue-Li Yang
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jian-Xin Li
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jie Cao
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xiang-Feng Lu
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jian-Feng Huang
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Ying Li
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Ji-Chun Chen
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Lian-Cheng Zhao
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Chong Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dong-Sheng Hu
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong 518071, China
| | - Ying-Xin Zhao
- Shandong First Medical University, Jinan, Shandong 271000, China
| | - Ling Yu
- Department of Cardiology, Fujian Provincial People's Hospital, Fuzhou, Fujian 350004, China
| | - Xiao-Qing Liu
- Division of Epidemiology, Guangdong Provincial People's Hospital and Cardiovascular Institute, Guangzhou, Guangdong 510080, China
| | - Xian-Ping Wu
- Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan 610041, China
| | - Dong-Feng Gu
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
160
|
Mrudulakumari Vasudevan U, Lee EY. Flavonoids, terpenoids, and polyketide antibiotics: Role of glycosylation and biocatalytic tactics in engineering glycosylation. Biotechnol Adv 2020; 41:107550. [PMID: 32360984 DOI: 10.1016/j.biotechadv.2020.107550] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/19/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
Flavonoids, terpenoids, and polyketides are structurally diverse secondary metabolites used widely as pharmaceuticals and nutraceuticals. Most of these molecules exist in nature as glycosides, in which sugar residues act as a decisive factor in their architectural complexity and bioactivity. Engineering glycosylation through selective trimming or extension of the sugar residues in these molecules is a prerequisite to their commercial production as well to creating novel derivatives with specialized functions. Traditional chemical glycosylation methods are tedious and can offer only limited end-product diversity. New in vitro and in vivo biocatalytic tools have emerged as outstanding platforms for engineering glycosylation in these three classes of secondary metabolites to create a large repertoire of versatile glycoprofiles. As knowledge has increased about secondary metabolite-associated promiscuous glycosyltransferases and sugar biosynthetic machinery, along with phenomenal progress in combinatorial biosynthesis, reliable industrial production of unnatural secondary metabolites has gained momentum in recent years. This review highlights the significant role of sugar residues in naturally occurring flavonoids, terpenoids, and polyketide antibiotics. General biocatalytic tools used to alter the identity and pattern of sugar molecules are described, followed by a detailed illustration of diverse strategies used in the past decade to engineer glycosylation of these valuable metabolites, exemplified with commercialized products and patents. By addressing the challenges involved in current bio catalytic methods and considering the perspectives portrayed in this review, exceptional drugs, flavors, and aromas from these small molecules could come to dominate the natural-product industry.
Collapse
Affiliation(s)
| | - Eun Yeol Lee
- Department of Chemical Engineering, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
161
|
Hirahatake KM, Astrup A, Hill JO, Slavin JL, Allison DB, Maki KC. Potential Cardiometabolic Health Benefits of Full-Fat Dairy: The Evidence Base. Adv Nutr 2020; 11:533-547. [PMID: 31904812 PMCID: PMC7231591 DOI: 10.1093/advances/nmz132] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/17/2019] [Accepted: 12/09/2019] [Indexed: 12/20/2022] Open
Abstract
Since their inception in 1980, the Dietary Guidelines for Americans have promoted low- or fat-free dairy foods. Removing fat from dairy does not reduce putatively beneficial nutrients per serving, including calcium, vitamin D, and potassium. Additionally, links between saturated fat and dietary cholesterol intakes with cardiovascular disease risk have helped to sustain the view that low-fat dairy foods should be recommended. Emerging evidence shows that the consumption of full-fat dairy foods has a neutral or inverse association with adverse cardiometabolic health outcomes, including atherosclerotic cardiovascular disease, type 2 diabetes, and associated risk factors. Thus, although low-fat dairy is a practical, practice-based recommendation, its superiority compared with full-fat dairy is not obviously supported by results from recent prospective cohort studies or intervention trials. To evaluate the emerging science on full-fat dairy, a group of nutrition experts convened to summarize and discuss the scientific evidence regarding the health effects of consuming full-fat dairy foods. Future studies should focus on full-fat dairy foods (milk, yogurt, and cheese) in the context of recommended dietary patterns and consider meal composition and metabolic phenotype in assessing the relation between full-fat dairy consumption and cardiometabolic health.
Collapse
Affiliation(s)
- Kristin M Hirahatake
- Department of Epidemiology, College of Health Sciences, University of California, Irvine, Irvine, CA, USA
| | - Arne Astrup
- Department of Nutrition, Exercise, and Sports, Copenhagen University, Copenhagen, Denmark
| | - James O Hill
- Center for Human Nutrition, University of Colorado School of Medicine, Denver, CO, USA
| | - Joanne L Slavin
- Department of Food Science and Nutrition, University of Minnesota, St Paul, MN, USA
| | - David B Allison
- School of Public Health, Indiana University, Bloomington, IN, USA
| | - Kevin C Maki
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, USA,Midwest Biomedical Research, Center for Metabolic and Cardiovascular Health, Addison, IL, USA,Address correspondence to KCM (e-mail: )
| |
Collapse
|
162
|
Matrisciano F, Pinna G. PPAR and functional foods: Rationale for natural neurosteroid-based interventions for postpartum depression. Neurobiol Stress 2020; 12:100222. [PMID: 32426424 PMCID: PMC7226878 DOI: 10.1016/j.ynstr.2020.100222] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/23/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023] Open
Abstract
Allopregnanolone, a GABAergic neurosteroid and progesterone derivative, was recently approved by the Food and Drug Administration for the treatment of postpartum depression (PPD). Several mechanisms appear to be involved in the pathogenesis of PPD, including neuroendocrine dysfunction, neuroinflammation, neurotransmitter alterations, genetic and epigenetic modifications. Recent evidence highlights the higher risk for incidence of PPD in mothers exposed to unhealthy diets that negatively impact the microbiome composition and increase inflammation, all effects that are strongly correlated with mood disorders. Conversely, healthy diets have consistently been reported to decrease the risk of peripartum depression and to protect the body and brain against low-grade systemic chronic inflammation. Several bioactive micronutrients found in the so-called functional foods have been shown to play a relevant role in preventing neuroinflammation and depression, such as vitamins, minerals, omega-3 fatty acids and flavonoids. An intriguing molecular substrate linking functional foods with improvement of mood disorders may be represented by the peroxisome-proliferator activated receptor (PPAR) pathway, which can regulate allopregnanolone biosynthesis and brain-derived neurotropic factor (BDNF) and thereby may reduce inflammation and elevate mood. Herein, we discuss the potential connection between functional foods and PPAR and their role in preventing neuroinflammation and symptoms of PPD through neurosteroid regulation. We suggest that healthy diets by targeting the PPAR-neurosteroid axis and thereby decreasing inflammation may offer a suitable functional strategy to prevent and safely alleviate mood symptoms during the perinatal period.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA
| |
Collapse
|
163
|
Flavonoids Distinctly Stabilize Lymph Endothelial- or Blood Endothelial Disintegration Induced by Colon Cancer Spheroids SW620. Molecules 2020; 25:molecules25092066. [PMID: 32365473 PMCID: PMC7248751 DOI: 10.3390/molecules25092066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/28/2020] [Indexed: 01/27/2023] Open
Abstract
The health effects of plant phenolics in vegetables and other food and the increasing evidence of the preventive potential of flavonoids in “Western Diseases” such as cancer, neurodegenerative diseases and others, have gained enormous interest. This prompted us to investigate the effects of 20 different flavonoids of the groups of flavones, flavonols and flavanones in 3D in vitro systems to determine their ability to inhibit the formation of circular chemorepellent induced defects (CCIDs) in monolayers of lymph- or blood-endothelial cells (LECs, BECs; respectively) by 12(S)-HETE, which is secreted by SW620 colon cancer spheroids. Several compounds reduced the spheroid-induced defects of the endothelial barriers. In the SW620/LEC model, apigenin and luteolin were most active and acacetin, nepetin, wogonin, pinocembrin, chrysin and hispidulin showed weak effects. In the SW620/BEC model acacetin, apigenin, luteolin, wogonin, hispidulin and chrysin exhibited weak activity.
Collapse
|
164
|
Rodriguez-Mateos A, Istas G, Boschek L, Feliciano RP, Mills CE, Boby C, Gomez-Alonso S, Milenkovic D, Heiss C. Circulating Anthocyanin Metabolites Mediate Vascular Benefits of Blueberries: Insights From Randomized Controlled Trials, Metabolomics, and Nutrigenomics. J Gerontol A Biol Sci Med Sci 2020; 74:967-976. [PMID: 30772905 DOI: 10.1093/gerona/glz047] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Indexed: 01/07/2023] Open
Abstract
Potential health benefits of blueberries may be due to vascular effects of anthocyanins that predominantly circulate in blood as phenolic acid metabolites. We investigated which role blueberry anthocyanins and circulating metabolites play in mediating improvements in vascular function and explore potential mechanisms using metabolomics and nutrigenomics. Purified anthocyanins exerted a dose-dependent improvement of endothelial function in healthy humans, as measured by flow-mediated dilation. The effects were similar to those of wild blueberries containing similar amounts of anthocyanins, whereas control drinks containing fiber, minerals, or vitamins had no significant effect. Daily 1-month wild blueberry consumption increased flow-mediated dilation and lowered 24-hour ambulatory systolic blood pressure. Of the 63 anthocyanin plasma metabolites quantified, 14 and 21 correlated with acute and chronic flow-mediated dilation improvements, respectively. Injection of these metabolites improved flow-mediated dilation in mice. Daily wild blueberry consumption led to differential expression (>1.2-fold) of 608 genes and 3 microRNAs, with Mir-181c showing a 13-fold increase in peripheral blood mononuclear cells. Patterns of 13 metabolites were independent predictors of gene expression changes and pathway enrichment analysis revealed significantly modulated biological processes involved in cell adhesion, migration, immune response, and cell differentiation. Our results identify anthocyanin metabolites as major mediators of vascular bioactivities of blueberries and changes of cellular gene programs. Trial registration: NCT025208.
Collapse
Affiliation(s)
- Ana Rodriguez-Mateos
- Department of Nutritional Sciences, School of Life Course Sciences, King's College London, UK.,Division of Cardiology, Pulmonology, and Vascular Medicine, University Düsseldorf, Germany
| | - Geoffrey Istas
- Department of Nutritional Sciences, School of Life Course Sciences, King's College London, UK.,Division of Cardiology, Pulmonology, and Vascular Medicine, University Düsseldorf, Germany
| | - Lisa Boschek
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Düsseldorf, Germany
| | - Rodrigo P Feliciano
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Düsseldorf, Germany
| | - Charlotte E Mills
- Department of Nutritional Sciences, School of Life Course Sciences, King's College London, UK.,Department of Food and Nutritional Sciences, University of Reading, UK
| | - Céline Boby
- Unité de Nutrition Humaine, INRA, UMR 1019, UNH, CRNH Auvergne, Clermont-Ferrand, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
| | - Sergio Gomez-Alonso
- Instituto Regional de Investigación Científica Aplicada, Universidad de Castilla-La Mancha, Spain
| | - Dragan Milenkovic
- Unité de Nutrition Humaine, INRA, UMR 1019, UNH, CRNH Auvergne, Clermont-Ferrand, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France.,Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis
| | - Christian Heiss
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Düsseldorf, Germany.,Faculty of Health and Medical Sciences, University of Surrey, Guildford.,Surrey and Sussex Healthcare NHS Trust, Redhill, UK
| |
Collapse
|
165
|
Bhavadharini B, Dehghan M, Mente A, Rangarajan S, Sheridan P, Mohan V, Iqbal R, Gupta R, Lear S, Wentzel-Viljoen E, Avezum A, Lopez-Jaramillo P, Mony P, Varma RP, Kumar R, Chifamba J, Alhabib KF, Mohammadifard N, Oguz A, Lanas F, Rozanska D, Bengtsson Bostrom K, Yusoff K, Tsolkile LP, Dans A, Yusufali A, Orlandini A, Poirier P, Khatib R, Hu B, Wei L, Yin L, Deeraili A, Yeates K, Yusuf R, Ismail N, Mozaffarian D, Teo K, Anand SS, Yusuf S. Association of dairy consumption with metabolic syndrome, hypertension and diabetes in 147 812 individuals from 21 countries. BMJ Open Diabetes Res Care 2020; 8:8/1/e000826. [PMID: 32423962 PMCID: PMC7326257 DOI: 10.1136/bmjdrc-2019-000826] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/07/2020] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Our aims were to assess the association of dairy intake with prevalence of metabolic syndrome (MetS) (cross-sectionally) and with incident hypertension and incident diabetes (prospectively) in a large multinational cohort study. METHODS The Prospective Urban Rural Epidemiology (PURE) study is a prospective epidemiological study of individuals aged 35 and 70 years from 21 countries on five continents, with a median follow-up of 9.1 years. In the cross-sectional analyses, we assessed the association of dairy intake with prevalent MetS and its components among individuals with information on the five MetS components (n=112 922). For the prospective analyses, we examined the association of dairy with incident hypertension (in 57 547 individuals free of hypertension) and diabetes (in 131 481 individuals free of diabetes). RESULTS In cross-sectional analysis, higher intake of total dairy (at least two servings/day compared with zero intake; OR 0.76, 95% CI 0.71 to 0.80, p-trend<0.0001) was associated with a lower prevalence of MetS after multivariable adjustment. Higher intakes of whole fat dairy consumed alone (OR 0.72, 95% CI 0.66 to 0.78, p-trend<0.0001), or consumed jointly with low fat dairy (OR 0.89, 95% CI 0.80 to 0.98, p-trend=0.0005), were associated with a lower MetS prevalence. Low fat dairy consumed alone was not associated with MetS (OR 1.03, 95% CI 0.77 to 1.38, p-trend=0.13). In prospective analysis, 13 640 people with incident hypertension and 5351 people with incident diabetes were recorded. Higher intake of total dairy (at least two servings/day vs zero serving/day) was associated with a lower incidence of hypertension (HR 0.89, 95% CI 0.82 to 0.97, p-trend=0.02) and diabetes (HR 0.88, 95% CI 0.76 to 1.02, p-trend=0.01). Directionally similar associations were found for whole fat dairy versus each outcome. CONCLUSIONS Higher intake of whole fat (but not low fat) dairy was associated with a lower prevalence of MetS and most of its component factors, and with a lower incidence of hypertension and diabetes. Our findings should be evaluated in large randomized trials of the effects of whole fat dairy on the risks of MetS, hypertension, and diabetes.
Collapse
Affiliation(s)
- Balaji Bhavadharini
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Mahshid Dehghan
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Andrew Mente
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Sumathy Rangarajan
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Patrick Sheridan
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Viswanathan Mohan
- Epidemiology, Madras Diabetes Research Foundation, Chennai, Tamil Nadu, India
- Diabetology, Dr Mohan's Diabetes Specialities Centre Gopalapuram, Chennai, Tamil Nadu, India
| | | | - Rajeev Gupta
- Preventive Cardiology, Eternal Heart Care Centre & Research Institute, Jaipur, Rajasthan, India
| | - Scott Lear
- Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Alvaro Avezum
- Research Division, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | | | - Prem Mony
- St John's Medical College & Research Institute, Bengaluru, Karnataka, India
| | - Ravi Prasad Varma
- Achutha Menon Centre for Health Science Studies, Thiruvananthapuram, Kerala, India
| | - Rajesh Kumar
- Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | | - Aytekin Oguz
- Internal Medicine, Goztepe Training and Research Hospital, Istanbul, Marmara, Turkey
| | | | | | | | - Khalid Yusoff
- Universiti Teknologi MARA, Shah Alam, Selangor, Malaysia
| | | | - Antonio Dans
- University of the Philippines System, Quezon City, Metro Manila, Philippines
| | | | | | - Paul Poirier
- Cardiology, Institut universitaire de cardiologie et de pneumologie de Quebec, Quebec City, Quebec, Canada
| | - Rasha Khatib
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bo Hu
- Peking Union Medical College School of Basic Medicine, Beijing, China
| | - Li Wei
- Medical Research & Biometrics Center, Chinese Academy of Medical Sciences and Peking Union Medical College Fuwai Hospital, Xicheng District, Beijing, China
| | - Lu Yin
- Peking Union Medical College School of Basic Medicine, Beijing, China
| | - Ai Deeraili
- Xinjiang Uighur Autonomous Region Center for Disease Control and Prevention, Wulumuqi, Xinjiang, China
| | | | - Rita Yusuf
- Independent University, Dhaka, Bangladesh
| | | | - Dariush Mozaffarian
- Tufts Friedman School of Nutrition Science & Policy, Boston, Massachusetts, USA
| | - Koon Teo
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sonia S Anand
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Salim Yusuf
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
166
|
Lechner K, von Schacky C, McKenzie AL, Worm N, Nixdorff U, Lechner B, Kränkel N, Halle M, Krauss RM, Scherr J. Lifestyle factors and high-risk atherosclerosis: Pathways and mechanisms beyond traditional risk factors. Eur J Prev Cardiol 2020; 27:394-406. [PMID: 31408370 PMCID: PMC7065445 DOI: 10.1177/2047487319869400] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/23/2019] [Indexed: 12/21/2022]
Abstract
Despite major efforts to reduce atherosclerotic cardiovascular disease (ASCVD) burden with conventional risk factor control, significant residual risk remains. Recent evidence on non-traditional determinants of cardiometabolic health has advanced our understanding of lifestyle-disease interactions. Chronic exposure to environmental stressors like poor diet quality, sedentarism, ambient air pollution and noise, sleep deprivation and psychosocial stress affect numerous traditional and non-traditional intermediary pathways related to ASCVD. These include body composition, cardiorespiratory fitness, muscle strength and functionality and the intestinal microbiome, which are increasingly recognized as major determinants of cardiovascular health. Evidence points to partially overlapping mechanisms, including effects on inflammatory and nutrient sensing pathways, endocrine signalling, autonomic function and autophagy. Of particular relevance is the potential of low-risk lifestyle factors to impact on plaque vulnerability through altered adipose tissue and skeletal muscle phenotype and secretome. Collectively, low-risk lifestyle factors cause a set of phenotypic adaptations shifting tissue cross-talk from a proinflammatory milieu conducive for high-risk atherosclerosis to an anti-atherogenic milieu. The ketone body ß-hydroxybutyrate, through inhibition of the NLRP-3 inflammasome, is likely to be an intermediary for many of these observed benefits. Adhering to low-risk lifestyle factors adds to the prognostic value of optimal risk factor management, and benefit occurs even when the impact on conventional risk markers is discouragingly minimal or not present. The aims of this review are (a) to discuss novel lifestyle risk factors and their underlying biochemical principles and (b) to provide new perspectives on potentially more feasible recommendations to improve long-term adherence to low-risk lifestyle factors.
Collapse
Affiliation(s)
- Katharina Lechner
- Technical University of Munich, School of Medicine, Department of Prevention, Rehabilitation and Sports Medicine, Germany
| | - Clemens von Schacky
- Preventive Cardiology, Ludwig-Maximilians University, Munich, Germany
- Omegametrix, Martinsried, Germany
| | | | - Nicolai Worm
- German University for Prevention and Health Care Management, Saarbrücken, Germany
| | - Uwe Nixdorff
- European Prevention Centre, Medical Centre Düsseldorf (Grand Arc), Germany
| | - Benjamin Lechner
- Department of Internal Medicine IV, Ludwig-Maximilians University, Munich, Germany
| | - Nicolle Kränkel
- Charité – Universitätsmedizin Berlin, Klinik für Kardiologie, Campus Benjamin Steglitz, Berlin, Germany
| | - Martin Halle
- Technical University of Munich, School of Medicine, Department of Prevention, Rehabilitation and Sports Medicine, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart Alliance, Germany
| | | | - Johannes Scherr
- Technical University of Munich, School of Medicine, Department of Prevention, Rehabilitation and Sports Medicine, Germany
- University Centre for Prevention and Sports Medicine, Balgrist University Hospital, University of Zurich, Switzerland
| |
Collapse
|
167
|
Ebbeling CB. Confusion at the milk cooler: opportunity to bolster the evidence base for preventive nutrition. Am J Clin Nutr 2020; 111:240-241. [PMID: 31901161 DOI: 10.1093/ajcn/nqz319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
168
|
Abstract
PURPOSE OF REVIEW Due to inconsistent findings on dairy consumption and CVD and all-cause mortality, we performed a narrative literature review to the current literature on dairy and its association with CVD. RECENT FINDINGS Due to their complex biochemistry, dairy consumption is a rather heterogeneous exposure. Multiple pathways have been proposed from dairy consumption and CVD. Current guidelines advocate for consumption of low-fat dairy products particularly milk, cheese, and yogurt, although the evidence for this is scant. Randomized clinical trials and large prospective studies on lipid-related cardiometabolic disease risk factors are consistent with results from most meta-analyses of prospective cohort studies, which suggest null or inverse relationship between CVD risk and mortality with dairy consumption although there is no clear dose response relationship. Most of current evidence suggests that dairy products are neutral or positive effect on human cardiovascular diseases.
Collapse
|
169
|
Caro-Ordieres T, Marín-Royo G, Opazo-Ríos L, Jiménez-Castilla L, Moreno JA, Gómez-Guerrero C, Egido J. The Coming Age of Flavonoids in the Treatment of Diabetic Complications. J Clin Med 2020; 9:jcm9020346. [PMID: 32012726 PMCID: PMC7074336 DOI: 10.3390/jcm9020346] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM), and its micro and macrovascular complications, is one of the biggest challenges for world public health. Despite overall improvement in prevention, diagnosis and treatment, its incidence is expected to continue increasing over the next years. Nowadays, finding therapies to prevent or retard the progression of diabetic complications remains an unmet need due to the complexity of mechanisms involved, which include inflammation, oxidative stress and angiogenesis, among others. Flavonoids are natural antioxidant compounds that have been shown to possess anti-diabetic properties. Moreover, increasing scientific evidence has demonstrated their potential anti-inflammatory and anti-oxidant effects. Consequently, the use of these compounds as anti-diabetic drugs has generated growing interest, as is reflected in the numerous in vitro and in vivo studies related to this field. Therefore, the aim of this review is to assess the recent pre-clinical and clinical research about the potential effect of flavonoids in the amelioration of diabetic complications. In brief, we provide updated information concerning the discrepancy between the numerous experimental studies supporting the efficacy of flavonoids on diabetic complications and the lack of appropriate and well-designed clinical trials. Due to the well-described beneficial effects on different mechanisms involved in diabetic complications, the excellent tolerability and low cost, future randomized controlled studies with compounds that have adequate bioavailability should be evaluated as add-on therapy on well-established anti-diabetic drugs.
Collapse
Affiliation(s)
- Teresa Caro-Ordieres
- Research Discovery and Innovation Department, FAES FARMA, S.A, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, 48940 Leioa (Bizkaia), Spain;
| | - Gema Marín-Royo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autonoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (G.M.-R.); (L.O.-R.); (L.J.-C.); (C.G.-G.)
| | - Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autonoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (G.M.-R.); (L.O.-R.); (L.J.-C.); (C.G.-G.)
| | - Luna Jiménez-Castilla
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autonoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (G.M.-R.); (L.O.-R.); (L.J.-C.); (C.G.-G.)
| | - Juan Antonio Moreno
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain;
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain
- Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Carmen Gómez-Guerrero
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autonoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (G.M.-R.); (L.O.-R.); (L.J.-C.); (C.G.-G.)
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autonoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (G.M.-R.); (L.O.-R.); (L.J.-C.); (C.G.-G.)
- Correspondence:
| |
Collapse
|
170
|
The association between milk consumption and the metabolic syndrome: a cross-sectional study of the residents of Suzhou, China and a meta-analysis. Br J Nutr 2020; 123:1013-1023. [PMID: 31964442 DOI: 10.1017/s0007114520000227] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The association between milk consumption and the metabolic syndrome remains inconclusive, and data from Chinese populations are scarce. We conducted a cross-sectional study to investigate the association between milk consumption and the metabolic syndrome and its components among the residents of Suzhou Industrial Park, Suzhou, China. A total of 5149 participants were included in the final analysis. A logistic regression model was applied to estimate the OR and 95 % CI for the prevalence of the metabolic syndrome and its components according to milk consumption. In addition, the results of our study were further meta-analysed with other published observational studies to quantify the association between the highest v. lowest categories of milk consumption and the metabolic syndrome and its components. There was no significant difference in the odds of having the metabolic syndrome between milk consumers and non-milk consumers (OR 0·86, 95 % CI 0·73, 1·01). However, milk consumers had lower odds of having elevated waist circumference (OR 0·78, 95 % CI 0·67, 0·92), elevated TAG (OR 0·83, 95 % CI 0·70, 0·99) and elevated blood pressure (OR 0·85, 95 % CI 0·73, 0·99). When the results were pooled together with other published studies, higher milk consumption was inversely associated with the risk of the metabolic syndrome (relative risk 0·80, 95 % CI 0·72, 0·88) and its components (except elevated fasting blood glucose); however, these results should be treated with caution as high heterogeneity was observed. In summary, the currently available evidence from observational studies suggests that higher milk consumption may be inversely associated with the metabolic syndrome.
Collapse
|
171
|
Abstract
The effect of dietary fats on cardiometabolic diseases, including cardiovascular diseases and type 2 diabetes mellitus, has generated tremendous interest. Many earlier investigations focused on total fat and conventional fat classes (such as saturated and unsaturated fats) and their influence on a limited number of risk factors. However, dietary fats comprise heterogeneous molecules with diverse structures, and growing research in the past two decades supports correspondingly complex health effects of individual dietary fats. Moreover, health effects of dietary fats might be modified by additional factors, such as accompanying nutrients and food-processing methods, emphasizing the importance of the food sources. Accordingly, the rapidly increasing scientific findings on dietary fats and cardiometabolic diseases have generated debate among scientists, caused confusion for the general public and present challenges for translation into dietary advice and policies. This Review summarizes the evidence on the effects of different dietary fats and their food sources on cell function and on risk factors and clinical events of cardiometabolic diseases. The aim is not to provide an exhaustive review but rather to focus on the most important evidence from randomized controlled trials and prospective cohort studies and to highlight current areas of controversy and the most relevant future research directions for understanding how to improve the prevention and management of cardiometabolic diseases through optimization of dietary fat intake.
Collapse
|
172
|
Mozaffarian D. Dietary and policy priorities to reduce the global crises of obesity and diabetes. ACTA ACUST UNITED AC 2020. [DOI: 10.1038/s43016-019-0013-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
173
|
Soedamah-Muthu SS, Guo J. Dairy consumption and cardiometabolic diseases: Evidence from prospective studies. MILK AND DAIRY FOODS 2020:1-28. [DOI: 10.1016/b978-0-12-815603-2.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
174
|
Zhou P, Hua F, Wang X, Huang JL. Therapeutic potential of IKK-β inhibitors from natural phenolics for inflammation in cardiovascular diseases. Inflammopharmacology 2020; 28:19-37. [PMID: 31894515 DOI: 10.1007/s10787-019-00680-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVDs) is a chronic disease with the highest morbidity and mortality in the world. Previous studies have suggested that preventing inflammation serves an efficient role in protection against cardiovascular diseases. Modulation of IKK-β activity can be used to treat and control CVDs associated with chronic inflammation, which targets the phosphorylation of IκB following the release of the RelA complex, and then translocates to the nucleus, eventually triggering the transcription of several genes that induce chemokines, cytokines, and adhesion molecules. Most importantly, the IκB kinase (IKK) complex is involved in transcriptional activation by phosphorylating the inhibitory molecule IkBα, enabling activation of NF-κB. Phenolic compounds possess cardioprotective potential that may be related to modulating inflammatory responses involved in CVDs. The SystemsDock analysis was used to explore whether 38 active compounds inhibit IKK-β activity based on literature. Docking results showed that the top docking score of three chemical compounds were icariin, salvianolic acid B, and plantainoside D in all compounds. Icariin, salvianolic acid B, and plantainoside D are the most promising IKKβ inhibitors. These phytochemicals could be helpful to find the lead compounds on designing and developing novel cardioprotective agents.
Collapse
Affiliation(s)
- Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China.
| | - Fang Hua
- Pharmacy School, Anhui Xinhua University, Hefei, 230088, People's Republic of China.,Natural Products Laboratory, International Joint Lab of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, 230036, People's Republic of China
| | - Xiang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China
| | - Jin-Ling Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China.
| |
Collapse
|
175
|
Thomas DD, Stockman MC, Yu L, Meshulam T, McCarthy AC, Ionson A, Burritt N, Deeney J, Cabral H, Corkey B, Istfan N, Apovian CM. Effects of medium chain triglycerides supplementation on insulin sensitivity and beta cell function: A feasibility study. PLoS One 2019; 14:e0226200. [PMID: 31869355 PMCID: PMC6927614 DOI: 10.1371/journal.pone.0226200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/20/2019] [Indexed: 12/20/2022] Open
Abstract
Objective Medium chain triglycerides (MCT) have unique metabolic properties which may improve insulin sensitivity (Si) and beta cell function but data in humans are limited. We conducted a 6-week clinical trial of MCT oil supplementation. Methods 22 subjects without diabetes (8 males, 14 females, mean ± standard error age 39±2.9 years, baseline BMI 27.0±1.4 kg/m2) were counseled to maintain their body weight and physical activity (PA) during the trial. Dietary intake, PA data, body composition, and resting energy expenditure (REE) were obtained through dietary recall, international PA questionnaire, dual x-ray absorptiometry, and indirect calorimetry, respectively. MCT prescriptions were given based on REE and PA to replace part of dietary fat with 30 grams of MCT per 2000 kcal daily. Insulin-modified frequently sampled intravenous glucose tolerance tests were performed before and after MCT to measure changes in Si, acute insulin response (AIR), disposition index (DI), and glucose effectiveness (Sg). Results MCT were well tolerated and weight remained stable (mean change 0.3 kg, p = 0.39). Fasting REE, respiratory quotient, and body composition were stable during the intervention. There were no significant changes in mean fasting glucose, insulin, insulin resistance, fasting total ketones, Si, AIR, DI, Sg, leptin, fructosamine, and proinsulin. The mean change in Si was 0.5 10−4 min-1 per mU/L (95% CI: -1.4, 2.4), corresponding to a 12% increase from baseline, and the range was -4.7 to 12.9 10−4 min-1 per mU/L. Mean total adiponectin decreased significantly from 22925 ng/mL at baseline to 17598 ng/mL at final visit (p = 0.02). The baseline clinical and laboratory parameters were not significantly associated with the change in Si. Discussion There were a wide range of changes in the minimal model parameters of glucose and insulin metabolism in subjects following 6 weeks of MCT as an isocaloric substitution for part of usual dietary fat intake. Since this was a single-arm non-randomized study without a control group, it cannot be certain whether these changes were due to MCT so further randomized controlled trials are warranted.
Collapse
Affiliation(s)
- Dylan D. Thomas
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Mary-Catherine Stockman
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Liqun Yu
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Tova Meshulam
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Ashley C. McCarthy
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Annaliese Ionson
- Boston University School of Medicine, Boston, MA, United States of America
| | - Nathan Burritt
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Jude Deeney
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Howard Cabral
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | - Barbara Corkey
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Nawfal Istfan
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Caroline M. Apovian
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
176
|
Morikawa T, Nagatomo A, Oka T, Miki Y, Taira N, Shibano-Kitahara M, Hori Y, Muraoka O, Ninomiya K. Glucose Tolerance-Improving Activity of Helichrysoside in Mice and Its Structural Requirements for Promoting Glucose and Lipid Metabolism. Int J Mol Sci 2019; 20:ijms20246322. [PMID: 31847420 PMCID: PMC6941121 DOI: 10.3390/ijms20246322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023] Open
Abstract
An acylated flavonol glycoside, helichrysoside, at a dose of 10 mg/kg/day per os for 14 days, improved the glucose tolerance in mice without affecting the food intake, visceral fat weight, liver weight, and other plasma parameters. In this study, using hepatoblastoma-derived HepG2 cells, helichrysoside, trans-tiliroside, and kaempferol 3-O-β-d-glucopyranoside enhanced glucose consumption from the medium, but their aglycones and p-coumaric acid did not show this activity. In addition, several acylated flavonol glycosides were synthesized to clarify the structural requirements for lipid metabolism using HepG2 cells. The results showed that helichrysoside and related analogs significantly inhibited triglyceride (TG) accumulation in these cells. The inhibition by helichrysoside was more potent than that by other acylated flavonol glycosides, related flavonol glycosides, and organic acids. As for the TG metabolism-promoting activity in high glucose-pretreated HepG2 cells, helichrysoside, related analogs, and their aglycones were found to significantly reduce the TG contents in HepG2 cells. However, the desacyl flavonol glycosides and organic acids derived from the acyl groups did not exhibit an inhibitory impact on the TG contents in HepG2 cells. These results suggest that the existence of the acyl moiety at the 6′′ position in the D-glucopyranosyl part is essential for glucose and lipid metabolism-promoting activities.
Collapse
Affiliation(s)
- Toshio Morikawa
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
- Antiaging Center, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan
- Correspondence: ; Tel.: +81-6-4307-4306; Fax: +81-6-6729-3577
| | - Akifumi Nagatomo
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
| | - Takahiro Oka
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
| | - Yoshinobu Miki
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
| | - Norihisa Taira
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
| | - Megumi Shibano-Kitahara
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
| | - Yuichiro Hori
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
| | - Osamu Muraoka
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
- Antiaging Center, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan
| | - Kiyofumi Ninomiya
- Pharmaceutical Research and Technology Institute, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan; (A.N.); (T.O.); (Y.M.); (N.T.); (M.S.-K.); (Y.H.); (O.M.); (K.N.)
- Antiaging Center, Kindai University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan
| |
Collapse
|
177
|
Carro A, Panisello JM. Deciphering the Riddles in Nutrition and Cardiovascular Disease. Eur Cardiol 2019; 14:141-150. [PMID: 31933681 PMCID: PMC6950215 DOI: 10.15420/ecr.2019.07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the leading global cause of death in Western countries, and its development is largely associated with unhealthy dietary patterns. A large body of scientific evidence has reported that nutrition might be the most preventive factor of cardiovascular disease death and could even reverse heart disease. Processes of chronic inflammation and oxidative distress share triggers that are modifiable by nutrition. This review aimed to identify potential targets (food patterns, single foods or individual nutrients) for cardiovascular disease prevention, and analyse the mechanisms implicated in their cardioprotective effects.
Collapse
|
178
|
Jia L, Zhao W, Sang J, Wang W, Wei W, Wang Y, Zhao F, Lu F, Liu F. Inhibitory Effect of a Flavonoid Dihydromyricetin against Aβ40 Amyloidogenesis and Its Associated Cytotoxicity. ACS Chem Neurosci 2019; 10:4696-4703. [PMID: 31596069 DOI: 10.1021/acschemneuro.9b00480] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Misfolding and fibrillogenesis of amyloid-β protein (Aβ) play a key role in the onset and progression of Alzheimer's disease (AD). Screening for inhibitors against Aβ amyloidogenesis is helpful for rational designing and developing new anti-AD drugs and therapeutic strategies. Dihydromyricetin, a natural flavonoid extracted from a Chinese herb, Ampelopsis grossedentata, has been proven with antioxidative, anti-inflammatory, and neuroprotective effects against neurodegenerative disease. Herein, we found that dihydromyricetin could inhibit Aβ40 aggregation, impede the protofibril formation, disassemble preformed Aβ40 fibrils, and protect PC12 cells from the Aβ40-induced cytotoxicity using a series of biochemical and biophysical assays, including thioflavin T fluorescence, atomic force microscopy, and cell toxicity assays. Circular dichroism spectroscopy data proved that dihydromyricetin delayed the Aβ40 conformational conversion. In addition, the results of molecular dynamics simulations indicated that the interaction between dihydromyricetin and Aβ40 trimer is mainly nonpolar interactions. Key residues (i.e., V18, A21, and D23) of the Aβ40 interacting with dihydromyricetin were also identified. This study suggested that dihydromyricetin shows great potential to be developed as a novel Aβ40 inhibitor.
Collapse
Affiliation(s)
- Longgang Jia
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Wenping Zhao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Jingcheng Sang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Wenjuan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Wei Wei
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Ying Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Fang Zhao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, P. R. China
- Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, P. R. China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| |
Collapse
|
179
|
7- O-methylpunctatin, a Novel Homoisoflavonoid, Inhibits Phenotypic Switch of Human Arteriolar Smooth Muscle Cells. Biomolecules 2019; 9:biom9110716. [PMID: 31717401 PMCID: PMC6920859 DOI: 10.3390/biom9110716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/09/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022] Open
Abstract
Remodeling of arterioles is a pivotal event in the manifestation of many inflammation-based cardio-vasculopathologies, such as hypertension. During these remodeling events, vascular smooth muscle cells (VSMCs) switch from a contractile to a synthetic phenotype. The latter is characterized by increased proliferation, migration, and invasion. Compounds with anti-inflammatory actions have been successful in attenuating this phenotypic switch. While the vast majority of studies investigating phenotypic modulation were undertaken in VSMCs isolated from large vessels, little is known about the effect of such compounds on phenotypic switch in VSMCs of microvessels (microVSMCs). We have recently characterized a novel homoisoflavonoid that we called 7-O-methylpunctatin (MP). In this study, we show that MP decreased FBS-induced cell proliferation, migration, invasion, and adhesion. MP also attenuated adhesion of THP-1 monocytes to microVSMCs, abolished FBS-induced expression of MMP-2, MMP-9, and NF-κB, as well as reduced activation of ERK1/2 and FAK. Furthermore, MP-treated VSMCs showed an increase in early (myocardin, SM-22α, SM-α) and mid-term (calponin and caldesmon) differentiation markers and a decrease in osteopontin, a protein highly expressed in synthetic VSMCs. MP also reduced transcription of cyclin D1, CDK4 but increased protein levels of p21 and p27. Taken together, these results corroborate an anti-inflammatory action of MP on human microVSMCs. Therefore, by inhibiting the synthetic phenotype of microVSMCs, MP may be a promising modulator for inflammation-induced arteriolar pathophysiology.
Collapse
|
180
|
Bilia AR, Piazzini V, Risaliti L, Vanti G, Casamonti M, Wang M, Bergonzi MC. Nanocarriers: A Successful Tool to Increase Solubility, Stability and Optimise Bioefficacy of Natural Constituents. Curr Med Chem 2019; 26:4631-4656. [PMID: 30381065 DOI: 10.2174/0929867325666181101110050] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/31/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
Abstract
Natural products are fascinating molecules in drug discovery for their exciting structure variability and also for their interaction with various targets. Drugs multi-targeting effect represents a more realistic approach to develop successful medications for many diseases. However, besides a large number of successful in vitro and in vivo studies, most of the clinical trials fail. This is generally related to the scarce water solubility, low lipophilicity and inappropriate molecular size of natural compounds, which undergo structural instability in biological milieu, rapid clearance and high metabolic rate. Additionally, some molecules are destroyed in gastric juice or suffer to a massive pre-systemic metabolism in the liver, when administered orally, limiting their clinical use. A reduced bioavailability can also be linked to drug distribution/accumulation in non-targeted tissues and organs that increase the side effects lowering the therapeutic efficacy and patient compliance. Nanomedicine represents a favourable tool to increase bioavailability and activities of natural products. Generally, nanovectors provide a large surface area and can overcome anatomic barriers. Each nanovector has its own advantages, disadvantages, and characteristics. In this review, different nanocarriers made of compounds which are Generally Recognized As Safe (GRAS) for the delivery of natural products, marketed as food supplements and medicines are reported.
Collapse
Affiliation(s)
- Anna Rita Bilia
- Department of Chemistry "Ugo Schiff", University of Florence, via Ugo Schiff 6, 50121, Sesto Fiorentino, Florence, Italy
| | - Vieri Piazzini
- Department of Chemistry "Ugo Schiff", University of Florence, via Ugo Schiff 6, 50121, Sesto Fiorentino, Florence, Italy
| | - Laura Risaliti
- Department of Chemistry "Ugo Schiff", University of Florence, via Ugo Schiff 6, 50121, Sesto Fiorentino, Florence, Italy
| | - Giulia Vanti
- Department of Chemistry "Ugo Schiff", University of Florence, via Ugo Schiff 6, 50121, Sesto Fiorentino, Florence, Italy
| | - Marta Casamonti
- Department of Chemistry "Ugo Schiff", University of Florence, via Ugo Schiff 6, 50121, Sesto Fiorentino, Florence, Italy
| | - Meng Wang
- Department of Chemistry "Ugo Schiff", University of Florence, via Ugo Schiff 6, 50121, Sesto Fiorentino, Florence, Italy.,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Yuquan Road 88, Tianjin, 300193, China
| | - Maria Camilla Bergonzi
- Department of Chemistry "Ugo Schiff", University of Florence, via Ugo Schiff 6, 50121, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
181
|
Drouin-Chartier JP, Li Y, Ardisson Korat AV, Ding M, Lamarche B, Manson JE, Rimm EB, Willett WC, Hu FB. Changes in dairy product consumption and risk of type 2 diabetes: results from 3 large prospective cohorts of US men and women. Am J Clin Nutr 2019; 110:1201-1212. [PMID: 31504094 PMCID: PMC6821541 DOI: 10.1093/ajcn/nqz180] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/10/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Whether changes in dairy product consumption are related to subsequent risk of type 2 diabetes (T2D) remains unknown. OBJECTIVE We evaluated the association of long-term changes in dairy product consumption with subsequent risk of T2D among US men and women. METHODS We followed up 34,224 men in the Health Professionals Follow-Up Study (1986-2012), 76,531 women in the Nurses' Health Study (1986-2012), and 81,597 women in the Nurses' Health Study II (1991-2013). Changes in dairy consumption were calculated from consecutive quadrennial FFQs. Multivariable Cox proportional regression models were used to calculate HRs for T2D associated with changes in dairy product consumption. Results of the 3 cohorts were pooled using an inverse variance-weighted, fixed-effect meta-analysis. RESULTS During 2,783,210 person-years, we documented 11,906 incident T2D cases. After adjustment for initial and changes in diet and lifestyle covariates, decreasing total dairy intake by >1.0 serving/d over a 4-y period was associated with an 11% (95% CI: 3%, 19%) higher risk of T2D in the subsequent 4 y compared with maintaining a relatively stable consumption (i.e., change in intake of ±1.0 serving/wk). Increasing yogurt consumption by >0.5 serving/d was associated with an 11% (95% CI: 4%, 18%) lower T2D risk, whereas increasing cheese consumption by >0.5 serving/d was associated with a 9% (95% CI: 2%, 16%) higher risk compared with maintaining stable intakes. Substituting 1 serving/d of yogurt or reduced-fat milk for cheese was associated with a 16% (95% CI: 10%, 22%) or 12% (95% CI: 8%, 16%) lower T2D risk, respectively. CONCLUSIONS Increasing yogurt consumption was associated with a moderately lower risk of T2D, whereas increasing cheese consumption was associated with a moderately higher risk among US men and women. Our study suggests that substituting yogurt or reduced-fat milk for cheese is associated with a lower risk of T2D.
Collapse
Affiliation(s)
- Jean-Philippe Drouin-Chartier
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Address correspondence to J-PD-C e-mail:
| | - Yanping Li
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Andres Victor Ardisson Korat
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ming Ding
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Benoît Lamarche
- School of Nutrition and Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - JoAnn E Manson
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA,Address correspondence to FBH e-mail:
| |
Collapse
|
182
|
Mozaffarian D. Dairy foods, dairy fat, diabetes, and death: what can be learned from 3 large new investigations? Am J Clin Nutr 2019; 110:1053-1054. [PMID: 31562511 PMCID: PMC6821539 DOI: 10.1093/ajcn/nqz250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
183
|
Guo J, Givens DI, Astrup A, Bakker SJL, Goossens GH, Kratz M, Marette A, Pijl H, Soedamah‐Muthu SS. The Impact of Dairy Products in the Development of Type 2 Diabetes: Where Does the Evidence Stand in 2019? Adv Nutr 2019; 10:1066-1075. [PMID: 31124561 PMCID: PMC6855942 DOI: 10.1093/advances/nmz050] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/26/2019] [Accepted: 04/18/2019] [Indexed: 12/21/2022] Open
Abstract
The prevalence of type 2 diabetes (T2D) has increased rapidly. Adopting a heathy diet is suggested as one of the effective behaviors to prevent or delay onset of T2D. Dairy consumption has been recommended as part of a healthy diet, but there remains uncertainty in both the scientific community and the public about the effect of different dairy products on T2D risk. In a recent workshop, the evidence on dairy products and T2D risk was presented and discussed by a group of experts. The main conclusions from the workshop are presented in this position paper and are as follows. 1) Available evidence from large prospective cohort studies and limited randomized controlled trials (RCTs) suggests that total dairy consumption has a neutral or moderately beneficial effect on T2D risk. 2) Increasing evidence from prospective cohort studies indicates that yogurt is most strongly associated with a lower T2D risk, but evidence from RCTs is scarce. 3) Fatty acids from dairy (medium-chain, odd, and very long-chain SFAs as well as trans-palmitoleic acid) are associated with lower T2D risk and improved metabolic health, but more research is needed on studies that explore cause and effect relations to exclude the possibility that the dairy fatty acids simply serve as markers of overall dairy consumption. 4) The food matrix can be a stronger determinant of health effects than SFA content. This review further identifies research gaps in the existing knowledge and highlights key research questions that need to be addressed to better understand the impact of dairy consumption on future T2D risk.
Collapse
Affiliation(s)
- Jing Guo
- Institute for Food, Nutrition and Health, University of Reading, Reading, United Kingdom
| | - D Ian Givens
- Institute for Food, Nutrition and Health, University of Reading, Reading, United Kingdom
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Mario Kratz
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - André Marette
- Faculty of Medicine, Laval University, Quebec, Canada
| | - Hanno Pijl
- Section of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Sabita S Soedamah‐Muthu
- Institute for Food, Nutrition and Health, University of Reading, Reading, United Kingdom
- Department of Medical and Clinical Psychology, Center of Research on Psychology in Somatic Diseases, Tilburg University, Tilburg, Netherlands
| |
Collapse
|
184
|
Park H, Jin UH, Orr AA, Echegaray SP, Davidson LA, Allred CD, Chapkin RS, Jayaraman A, Lee K, Tamamis P, Safe S. Isoflavones as Ah Receptor Agonists in Colon-Derived Cell Lines: Structure-Activity Relationships. Chem Res Toxicol 2019; 32:2353-2364. [PMID: 31621310 DOI: 10.1021/acs.chemrestox.9b00352] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many of the protective responses observed for flavonoids in the gastrointestinal track resemble aryl hydrocarbon receptor (AhR)-mediated effects. Therefore, we examined the structure-activity relationships of isoflavones and isomeric flavone and flavanones as AhR ligands on the basis of their induction of CYP1A1, CYP1B1, and UGT1A1 gene expression in colon cancer Caco2 cells and young adult mouse colonocyte (YAMC) cells. Caco2 cells were significantly more Ah-responsive than YAMC cells, and this was due, in part, to flavonoid-induced cytotoxicity in the latter cell lines. The structure-activity relationships for the flavonoids were complex and both response and cell context specific; however, there was significant variability in the AhR activities of the isomeric substituted isoflavones and flavones. For example, 4',5,7-trihydroxyisoflavone (genistein) was AhR-inactive whereas 4',5,7-trihydroxyflavone (apigenin) induced CYP1A1, CYP1B1, and UGT1A1 in Caco2 cells. In contrast, both 5,7-dihydroxy-4-methoxy substituted isoflavone (biochanin A) and flavone (acacetin) induced all three AhR-responsive genes; 4',5,7-trimethoxyisoflavone was a potent AhR agonist, and the isomeric flavone was AhR-inactive. These results coupled with simulation studies modeling flavonoid interaction within the AhR binding pocket demonstrate that the orientation of the substituted phenyl ring at C-2 (flavones) or C-3 (isoflavones) on the common 4-H-chromen-4-one ring strongly influences the activities of isoflavones and flavones as AhR agonists.
Collapse
Affiliation(s)
- Hyejin Park
- Department of Veterinary Physiology and Pharmacology , Texas A&M University , College Station , Texas 77843 , United States
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology , Texas A&M University , College Station , Texas 77843 , United States
| | - Asuka A Orr
- Artie McFerrin Department of Chemical Engineering , Texas A&M University , College Station , Texas 77840 , United States
| | - Stephanie P Echegaray
- Artie McFerrin Department of Chemical Engineering , Texas A&M University , College Station , Texas 77840 , United States
| | - Laurie A Davidson
- Department of Nutrition and Food Science , Texas A&M University , College Station , Texas 77843 , United States
| | - Clinton D Allred
- Department of Nutrition and Food Science , Texas A&M University , College Station , Texas 77843 , United States
| | - Robert S Chapkin
- Department of Nutrition and Food Science , Texas A&M University , College Station , Texas 77843 , United States
| | - Arul Jayaraman
- Artie McFerrin Department of Chemical Engineering , Texas A&M University , College Station , Texas 77840 , United States
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering , Tufts University , Medford , Massachusetts 02155 , United States
| | - Phanourios Tamamis
- Artie McFerrin Department of Chemical Engineering , Texas A&M University , College Station , Texas 77840 , United States
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology , Texas A&M University , College Station , Texas 77843 , United States
| |
Collapse
|
185
|
Contribution of plant food bioactives in promoting health effects of plant foods: why look at interindividual variability? Eur J Nutr 2019; 58:13-19. [PMID: 31637469 PMCID: PMC6851219 DOI: 10.1007/s00394-019-02096-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Purpose Research has identified plant-based diets as the most protective for our health; it is now essential to focus on good food associations and the beneficial constituents in plant foods. From a growing body of evidence, some categories of food phytochemicals are increasingly considered to play a crucial role in the cardiometabolic health effects associated with plant food consumption. However, the heterogeneity in responsiveness to plant food bioactive intake that is frequently observed in clinical trials can hinder the identification of the effects of these compounds in specific subpopulations and likely lead to underestimating their actual contribution to the health effects of their food sources. Results The magnitude and the main factors responsible for this between-subject variation in response to the consumption of the major families of food phytochemicals have been poorly documented so far. Thus, research efforts in this area must be developed. More importantly, capturing the interindividual variability in response to plant food bioactive intake, together with identifying the main determinants involved, is a crucial step that will enable the development and production of plant food products, thereby satisfying the nutritional needs and conferring benefits to different categories of populations. Conclusion The development of a science-based personalised nutrition approach focusing on plant foods rich in specific bioactive compounds could contribute to alleviating the dramatic burden of metabolic and cardiovascular diseases. Plant food bioactives represent a tremendous potential for innovation in the field of food, nutrition, and health. The role of phytochemicals in the health benefits of plant-based foods has been underestimated so far. A full consideration of plant food bioactives in future preventive strategies implies research that tackles the factors responsible for interindividual variability. A better understanding of interindividual variability in response to plant food bioactives is needed to:Refine dietary recommendations towards foods that are particularly rich in specific bioactives. Broaden perspectives for the food industry to develop strategies for future personalised food products targeted to specific consumer groups.
Collapse
|
186
|
Sinegre T, Teissandier D, Milenkovic D, Morand C, Lebreton A. Epicatechin influences primary hemostasis, coagulation and fibrinolysis. Food Funct 2019; 10:7291-7298. [PMID: 31621731 DOI: 10.1039/c9fo00816k] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The different stages of hemostasis (i.e., primary hemostasis, coagulation and fibrinolysis) are involved in the early atherothrombosis steps. The aim of this study was to investigate the effect of epicatechin, a major flavonoid compound, on the hemostasis phenotype using clinically relevant in vitro global assays that mimic the complexity of the in vivo hemostasis systems. Plasma samples from 10 healthy volunteers were spiked with increasing concentrations of epicatechin (1 to 100 μM). Epicatechin effect on primary hemostasis, coagulation and fibrinolysis was assessed by measuring platelet aggregation using light transmission aggregometry, thrombin generation and clot lysis time (CLT), respectively. Epicatechin (100 μM) significantly decreased the maximal platelet aggregation induced by adenosine diphosphate (-39%), thrombin receptor activating peptide (-48%), epinephrine (-30%), and collagen (-30%). The endogenous thrombin potential was significantly reduced starting from 1 μM epicatechin (1332 ± 230 versus 1548 ± 241 nM min for control) (p < 0.01). Fibrinolysis was promoted by epicatechin, as indicated by CLT decrease by 16 and 33% with 10 and 100 μM epicatechin respectively, compared with control (1271 ± 775 s). These findings show that epicatechin reduces platelet function and leads to an anticoagulant and pro-fibrinolytic profile, providing new evidence of its interest for cardiovascular disease prevention.
Collapse
Affiliation(s)
- Thomas Sinegre
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France. and CHU Clermont-Ferrand, service d'hématologie biologique, Clermont-Ferrand, France
| | - Dorian Teissandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France.
| | - Dragan Milenkovic
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France.
| | - Christine Morand
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France.
| | - Aurélien Lebreton
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France. and CHU Clermont-Ferrand, service d'hématologie biologique, Clermont-Ferrand, France
| |
Collapse
|
187
|
Castaldo L, Narváez A, Izzo L, Graziani G, Gaspari A, Di Minno G, Ritieni A. Red Wine Consumption and Cardiovascular Health. Molecules 2019; 24:E3626. [PMID: 31597344 PMCID: PMC6804046 DOI: 10.3390/molecules24193626] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/01/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023] Open
Abstract
Wine is a popular alcoholic beverage that has been consumed for hundreds of years. Benefits from moderate alcohol consumption have been widely supported by the scientific literature and, in this line, red wine intake has been related to a lesser risk for coronary heart disease (CHD). Experimental studies and meta-analyses have mainly attributed this outcome to the presence in red wine of a great variety of polyphenolic compounds such as resveratrol, catechin, epicatechin, quercetin, and anthocyanin. Resveratrol is considered the most effective wine compound with respect to the prevention of CHD because of its antioxidant properties. The mechanisms responsible for its putative cardioprotective effects would include changes in lipid profiles, reduction of insulin resistance, and decrease in oxidative stress of low-density lipoprotein cholesterol (LDL-C). The aim of this review is to summarize the accumulated evidence correlating moderate red wine consumption with prevention of CHD by focusing on the different mechanisms underlying this relationship. Furthermore, the chemistry of wine as well as chemical factors that influence the composition of the bioactive components of red wine are also discussed.
Collapse
Affiliation(s)
- Luigi Castaldo
- Department of Pharmacy, Faculty of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (L.C.); (A.N.); (L.I.); (G.G.); (A.G.)
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy;
| | - Alfonso Narváez
- Department of Pharmacy, Faculty of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (L.C.); (A.N.); (L.I.); (G.G.); (A.G.)
| | - Luana Izzo
- Department of Pharmacy, Faculty of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (L.C.); (A.N.); (L.I.); (G.G.); (A.G.)
| | - Giulia Graziani
- Department of Pharmacy, Faculty of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (L.C.); (A.N.); (L.I.); (G.G.); (A.G.)
| | - Anna Gaspari
- Department of Pharmacy, Faculty of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (L.C.); (A.N.); (L.I.); (G.G.); (A.G.)
| | - Giovanni Di Minno
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy;
| | - Alberto Ritieni
- Department of Pharmacy, Faculty of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (L.C.); (A.N.); (L.I.); (G.G.); (A.G.)
| |
Collapse
|
188
|
Navarro SL, Tarkhan A, Shojaie A, Randolph TW, Gu H, Djukovic D, Osterbauer KJ, Hullar MA, Kratz M, Neuhouser ML, Lampe PD, Raftery D, Lampe JW. Plasma metabolomics profiles suggest beneficial effects of a low-glycemic load dietary pattern on inflammation and energy metabolism. Am J Clin Nutr 2019; 110:984-992. [PMID: 31432072 PMCID: PMC6766441 DOI: 10.1093/ajcn/nqz169] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Low-glycemic load dietary patterns, characterized by consumption of whole grains, legumes, fruits, and vegetables, are associated with reduced risk of several chronic diseases. METHODS Using samples from a randomized, controlled, crossover feeding trial, we evaluated the effects on metabolic profiles of a low-glycemic whole-grain dietary pattern (WG) compared with a dietary pattern high in refined grains and added sugars (RG) for 28 d. LC-MS-based targeted metabolomics analysis was performed on fasting plasma samples from 80 healthy participants (n = 40 men, n = 40 women) aged 18-45 y. Linear mixed models were used to evaluate differences in response between diets for individual metabolites. Kyoto Encyclopedia of Genes and Genomes (KEGG)-defined pathways and 2 novel data-driven analyses were conducted to consider differences at the pathway level. RESULTS There were 121 metabolites with detectable signal in >98% of all plasma samples. Eighteen metabolites were significantly different between diets at day 28 [false discovery rate (FDR) < 0.05]. Inositol, hydroxyphenylpyruvate, citrulline, ornithine, 13-hydroxyoctadecadienoic acid, glutamine, and oxaloacetate were higher after the WG diet than after the RG diet, whereas melatonin, betaine, creatine, acetylcholine, aspartate, hydroxyproline, methylhistidine, tryptophan, cystamine, carnitine, and trimethylamine were lower. Analyses using KEGG-defined pathways revealed statistically significant differences in tryptophan metabolism between diets, with kynurenine and melatonin positively associated with serum C-reactive protein concentrations. Novel data-driven methods at the metabolite and network levels found correlations among metabolites involved in branched-chain amino acid (BCAA) degradation, trimethylamine-N-oxide production, and β oxidation of fatty acids (FDR < 0.1) that differed between diets, with more favorable metabolic profiles detected after the WG diet. Higher BCAAs and trimethylamine were positively associated with homeostasis model assessment-insulin resistance. CONCLUSIONS These exploratory metabolomics results support beneficial effects of a low-glycemic load dietary pattern characterized by whole grains, legumes, fruits, and vegetables, compared with a diet high in refined grains and added sugars on inflammation and energy metabolism pathways. This trial was registered at clinicaltrials.gov as NCT00622661.
Collapse
Affiliation(s)
- Sandi L Navarro
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Address correspondence to SLN (e-mail: )
| | - Aliasghar Tarkhan
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Ali Shojaie
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Timothy W Randolph
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Danijel Djukovic
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Katie J Osterbauer
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Meredith A Hullar
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mario Kratz
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marian L Neuhouser
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Paul D Lampe
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Daniel Raftery
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Johanna W Lampe
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
189
|
Astrup A, Geiker NRW, Magkos F. Effects of Full-Fat and Fermented Dairy Products on Cardiometabolic Disease: Food Is More Than the Sum of Its Parts. Adv Nutr 2019; 10:924S-930S. [PMID: 31518411 PMCID: PMC6743821 DOI: 10.1093/advances/nmz069] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/26/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
Current dietary recommendations to limit consumption of saturated fat are largely based on early nutrition studies demonstrating a direct link between dietary saturated fat, elevated blood cholesterol levels, and increased risk of cardiovascular disease. As full-fat dairy products are rich in saturated fat, these dietary guidelines recommend consumption of fat-free or low-fat dairy products in place of full-fat dairy. However, dairy products vary greatly in both their nutrient content and their bioactive ingredients, and research increasingly highlights the importance of focusing on whole foods (i.e., the food matrix) as opposed to single nutrients, such as saturated fat. In fact, the weight of evidence from recent large and well-controlled studies, systematic reviews, and meta-analyses of both observational studies and randomized controlled trials indicates that full-fat dairy products, particularly yogurt and cheese, do not exert the detrimental effects on insulin sensitivity, blood lipid profile, and blood pressure as previously predicted on the basis of their sodium and saturated fat contents; they do not increase cardiometabolic disease risk and may in fact protect against cardiovascular disease and type 2 diabetes. Although more research is warranted to adjust for possible confounding factors and to better understand the mechanisms of action of dairy products on health outcomes, it becomes increasingly clear that the recommendation to restrict dietary saturated fat to reduce risk of cardiometabolic disease is getting outdated. Therefore, the suggestion to restrict or eliminate full-fat dairy from the diet may not be the optimal strategy for reducing cardiometabolic disease risk and should be re-evaluated in light of recent evidence.
Collapse
Affiliation(s)
- Arne Astrup
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Nørre Campus, Copenhagen, Denmark
| | - Nina Rica Wium Geiker
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Frederiksberg Campus, Frederiksberg, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Frederiksberg Campus, Frederiksberg, Denmark
| |
Collapse
|
190
|
Mozaffarian D. Dairy Foods, Obesity, and Metabolic Health: The Role of the Food Matrix Compared with Single Nutrients. Adv Nutr 2019; 10:917S-923S. [PMID: 31518410 PMCID: PMC6743828 DOI: 10.1093/advances/nmz053] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/26/2019] [Accepted: 05/02/2019] [Indexed: 01/09/2023] Open
Abstract
In the 20th century, scientific and geopolitical events led to the concept of food as a delivery system for calories and specific isolated nutrients. As a result, conventional dietary guidelines have focused on individual nutrients to maintain health and prevent disease. For dairy foods, this has led to general dietary recommendations to consume 2-3 daily servings of reduced-fat dairy foods, without regard to type (e.g., yogurt, cheese, milk), largely based on theorized benefits of isolated nutrients for bone health (e.g., calcium, vitamin D) and theorized harms of isolated nutrients for cardiovascular diseases (CVDs) and obesity (e.g., total fat, saturated fat, total calories). However, advances in nutrition science have demonstrated that foods represent complex matrices of nutrients, minerals, bioactives, food structures, and other factors (e.g., phoshopholipids, prebiotics, probiotics) with correspondingly complex effects on health and disease. The present evidence suggests that whole-fat dairy foods do not cause weight gain, that overall dairy consumption increases lean body mass and reduces body fat, that yogurt consumption and probiotics reduce weight gain, that fermented dairy consumption including cheese is linked to lower CVD risk, and that yogurt, cheese, and even dairy fat may protect against type 2 diabetes. Based on the current science, dairy consumption is part of a healthy diet, without strong evidence to favor reduced-fat products; while intakes of probiotic-containing unsweetened and fermented dairy products such as yogurt and cheese appear especially beneficial.
Collapse
Affiliation(s)
- Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
191
|
Yang T, Feng YL, Chen L, Vaziri ND, Zhao YY. Dietary natural flavonoids treating cancer by targeting aryl hydrocarbon receptor. Crit Rev Toxicol 2019; 49:445-460. [PMID: 31433724 DOI: 10.1080/10408444.2019.1635987] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The role of aryl hydrocarbon receptor (AhR) as a ligand-activated transcription factor in the field of cancer has gradually been unveiled. A strong body of evidence indicated that AhR is implicated in cell proliferation and apoptosis, immune metabolism and other processes, which further affected tumor growth, survival, migration, and invasion. Therefore, AhR targeted therapy may become a new method for cancer treatment and provide a new direction for clinical tumor treatment. Astonishingly, the largest source of exposure of animals and humans to AhR ligands (synthetic and natural) comes from the diet. Myriad studies have described that various natural dietary chemicals can directly activate and/or inhibit the AhR signaling pathway. Of note, numerous natural products contribute to AhR active, of which dietary flavonoids are the largest class of natural AhR ligands. As interest in AhR and its ligands increases, it seems sensible to summarize current research on these ligands. In this review, we highlight the role of AhR in tumorigenesis and focus on the double effect of AhR in cancer therapy. We explored the molecular mechanism of AhR ligands on cancer through a few AhR agonists/antagonists currently in clinical practice. Ultimately, we summarize and highlight the latest progression of dietary flavonoids as AhR ligands in cancer inhibition, including the limitations and deficiencies of it in clinical research. This review will offer a comprehensive understanding of AhR and its dietary ligands which may dramatically pave the way for targeted cancer treatment.
Collapse
Affiliation(s)
- Tian Yang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Ya-Long Feng
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Lin Chen
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| |
Collapse
|
192
|
Ye Q, Liu K, Shen Q, Li Q, Hao J, Han F, Jiang RW. Reversal of Multidrug Resistance in Cancer by Multi-Functional Flavonoids. Front Oncol 2019; 9:487. [PMID: 31245292 PMCID: PMC6581719 DOI: 10.3389/fonc.2019.00487] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022] Open
Abstract
Multidrug resistance (MDR) resulting from different defensive mechanisms in cancer is one of the major obstacles of clinical treatment. To circumvent MDR many reversal agents have been developed, but most of them fail in clinical trials due to severely adverse effects. Recently, certain natural products have been reported to overcome MDR, including flavonoids which are abundant in plants, foods, and herbs. The structure of flavonoids can be abbreviated as C6-C3-C6 (C for carbon), and further categorized into flavonoids, iso-flavonoids and neo-flavonoids, according to their structural backbones. Flavonoids possess multiple bioactivities, and a growing body of research has indicated that both flavonoids and iso-flavonoids can either kill or re-sensitize conventional chemotherapeutics to resistant cancer cells. Here, we summarize the research and discuss the underlying mechanisms, concluding that these flavonoids do not function as specific regulators of target proteins, but rather as multi-functional agents that negatively regulate the key factors contributing to MDR.
Collapse
Affiliation(s)
| | - Kai Liu
- Hainan General Hospital, Haikou, China
| | - Qun Shen
- Hainan General Hospital, Haikou, China
| | | | - Jinghui Hao
- Jiaozuo Second People's Hospital, Jiaozuo, China
| | | | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
193
|
Coyle DH, Ndanuko R, Singh S, Huang P, Wu JH. Variations in Sugar Content of Flavored Milks and Yogurts: A Cross-Sectional Study across 3 Countries. Curr Dev Nutr 2019; 3:nzz060. [PMID: 31187086 PMCID: PMC6554456 DOI: 10.1093/cdn/nzz060] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/17/2019] [Accepted: 04/30/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The consumption of dairy products is encouraged at all life stages as a nutrient-rich component of the diet. However, many milk and yogurt products, particularly flavored varieties, may contain large amounts of free sugar. OBJECTIVES The aim of this paper was to evaluate the availability and sugar content of flavored milks and yogurts in supermarkets across 3 countries: Australia, England, and South. METHODS Nutrition information for flavored milks and yogurts was collected by trained researchers and supplemented by crowd-sourced data from a smartphone application. Data were extracted in April 2018 and 3724 milk and yogurt products were available for analysis. Mean sugar concentrations were compared across countries with the use of ANOVA followed by Tukey's post-hoc pairwise comparisons. Sugar concentrations were compared with the UK's "green" traffic-light classifications. RESULTS Approximately 74% (n = 2753) of all products were flavored. Flavored products contained nearly twice the average total sugar content of unflavored products, with substantial variability: mean total sugar was 9.1 g/100 mL (range: 4.3-15.0 g/100 mL) and 11.5 g/100 g (range: 0.1-22.6 g/100 g) for flavored milks and yogurts, respectively. Free sugars contributed an estimated 41% and 42% of total sugar in milks and yogurts, respectively. Flavored milks in England had ∼0.7 g/100 mL higher total sugar on average compared with Australia and South Africa (P ≤ 0.04), whereas flavored yogurts in South Africa had the lowest average total sugar (∼2 g/100 g lower than England and Australia; P < 0.001). Less than 4% of flavored products would receive a "green" rating under the UK traffic-light labeling scheme. CONCLUSIONS In Australia, England, and South Africa, flavored milks and yogurts are highly prevalent in the food supply and contain significantly higher concentrations of total and added sugars than unflavored products.
Collapse
Affiliation(s)
- Daisy H Coyle
- The George Institute for Global Health, Sydney, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| | - Rhoda Ndanuko
- The George Institute for Global Health, Sydney, NSW, Australia
| | - Sarinda Singh
- The George Institute for Global Health, Sydney, NSW, Australia
| | - Polly Huang
- The George Institute for Global Health, Sydney, NSW, Australia
| | - Jason H Wu
- The George Institute for Global Health, Sydney, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
194
|
Fantin M, Garelli F, Napoli B, Forgiarini A, Gumeni S, De Martin S, Montopoli M, Vantaggiato C, Orso G. Flavonoids Regulate Lipid Droplets Biogenesis in Drosophila melanogaster. Nat Prod Commun 2019. [DOI: 10.1177/1934578x19852430] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Lipid droplets (LDs), cytosolic fat storage organelles, are emerging as major regulators of lipid metabolism, trafficking, and signaling in various cells and tissues. LDs are altered in cardiovascular and neuronal disorders, inflammation, obesity, and cancer. Flavonoids comprise different classes of molecules, characterized by a well-known antioxidant activity and a beneficial effect in several diseases. However, the cellular mechanism by which different classes of flavonoids improve health is poorly understood, in particular as far as LDs biogenesis is concerned. Here we used Drosophila melanogaster as a model system to investigate the effects of a selected group of flavonoids on larval tissues by examining LDs biogenesis. In our study, fruit flies were grown in xanthohumol-, isoquercetin-, and genistein-enriched food and larval tissues were analyzed using a LD marker. Total mRNA expression of two main enzymes (minotaur and midway) responsible for triacylglycerides synthesis was evaluated after treatments. Among the flavonoids analyzed, xanthohumol and isoquercetin resulted to be potent regulators of LDs biogenesis in a tissue-specific manner, inducing fat storage decrease in fat bodies and accumulation of LDs in nerves. Since LDs have been suggested to play a protective role against intracellular stress in nonadipocyte cells, our data support the hypothesis that some phytochemicals could act as strong modulators of LDs biogenesis in vivo. The knowledge of how different flavonoids act on lipid metabolism in different tissues can help to manage the use of phytochemicals with the aim of selectively ameliorating specific neuronal and metabolic diseases’ manifestations.
Collapse
Affiliation(s)
- Marianna Fantin
- Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology, Bosisio Parini, Lecco, Italy
| | - Francesca Garelli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Barbara Napoli
- Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology, Bosisio Parini, Lecco, Italy
| | - Alessia Forgiarini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Chiara Vantaggiato
- Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology, Bosisio Parini, Lecco, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| |
Collapse
|
195
|
Lactose Intolerance and Bone Health: The Challenge of Ensuring Adequate Calcium Intake. Nutrients 2019; 11:nu11040718. [PMID: 30925689 PMCID: PMC6521087 DOI: 10.3390/nu11040718] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/21/2019] [Accepted: 03/23/2019] [Indexed: 02/07/2023] Open
Abstract
Calcium is an important nutrient with impact upon many biological systems, most notably bone. Ensuring adequate calcium intake throughout the lifespan is essential to building and maintaining bone. Lactose intolerance may predispose individuals to low calcium intake as the number of lactose-free, calcium-rich food sources is limited. In this review, we summarize data from human and animal studies on the influence of lactose and lactase deficiency on calcium absorption and bone health. Based on the available evidence, neither dietary lactose nor lactase deficiency have a significant impact on calcium absorption in adult humans. However, lactose intolerance may lead to reduced bone density and fragility fractures when accompanied by decreased intake or avoidance of dairy. Recently published human trials and meta-analyses suggest a weak but significant association between dairy consumption and bone health, particularly in children. Given the availability of simple dietary approaches to building lactose tolerance and the nutritional deficiencies associated with dairy avoidance, multiple public health organizations recommend that all individuals—including those that are lactose intolerant—consume three servings of dairy per day to ensure adequate nutrient intakes and optimal bone health.
Collapse
|
196
|
Guo K, Ren J, Gu G, Wang G, Gong W, Wu X, Ren H, Hong Z, Li J. Hesperidin Protects Against Intestinal Inflammation by Restoring Intestinal Barrier Function and Up-Regulating Treg Cells. Mol Nutr Food Res 2019; 63:e1800975. [PMID: 30817082 DOI: 10.1002/mnfr.201800975] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/20/2019] [Indexed: 12/19/2022]
Abstract
SCOPE Hesperidin is an important natural phenolic compound and is considered beneficial to health. The purpose of this study is to investigate the protective effects of hesperidin on DSS-induced colitis in mice and Caco-2 cells. METHODS The DSS-induced colitis mice are assigned to 10, 20, and 40 mg kg-g hesperidin diets after DSS treatment. For in vitro experiments, Caco-2 cells are treated with TNF-α/ IFN-γ for 48 h without or with hesperidin. RESULTS Hesperidin supplementation ameliorates DSS-induced colitis. Specifically, hesperidin ameliorates intestinal inflammation through decreasing MDA activity and enhancing SOD and GSH activities. Hesperidin also obviously upregulates Nrf2 antioxidant pathway and increases the protein expression of HO-1 and NQO1. Additionally, hesperidin significantly reduces the levels of inflammatory factors and increases the levels of anti-inflammatory factors in the colon tissues. Further analysis shows that hesperidin can improve the expression of tight junction proteins and intestinal permeability, as well as increases the Treg population. In Caco-2 cells, it is shown that hesperidin prevents TNF-α/IFN-γ-induced reduction in TEER and morphological disruption. Moreover, hesperidin also decreases the epithelial permeability and suppresses proinflammatory responses. CONCLUSION Hesperidin can protect against intestinal inflammation via enhanced Nrf2 antioxidant pathway, increases the Treg population, and restores intestinal barrier function.
Collapse
Affiliation(s)
- Kun Guo
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Jianan Ren
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Guosheng Gu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Gefei Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Wenbin Gong
- Department of General Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210002, China
| | - Xiuwen Wu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Huajian Ren
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Zhiwu Hong
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| |
Collapse
|
197
|
Madhurakkat Perikamana SK, Lee SM, Lee J, Ahmad T, Lee MS, Yang HS, Shin H. Oxidative Epigallocatechin Gallate Coating on Polymeric Substrates for Bone Tissue Regeneration. Macromol Biosci 2019; 19:e1800392. [DOI: 10.1002/mabi.201800392] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/21/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Sajeesh Kumar Madhurakkat Perikamana
- Department of BioengineeringHanyang University 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
| | - Sang Min Lee
- Department of BioengineeringHanyang University 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
| | - Jinkyu Lee
- Department of BioengineeringHanyang University 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
| | - Taufiq Ahmad
- Department of BioengineeringHanyang University 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
| | - Min Suk Lee
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook University Cheonan 31116 Republic of Korea
| | - Hee Seok Yang
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook University Cheonan 31116 Republic of Korea
| | - Heungsoo Shin
- Department of BioengineeringHanyang University 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team 222 Wangsimni‐ro Seongdong‐gu Seoul 04763 Republic of Korea
| |
Collapse
|
198
|
Kong F, Lee BH, Wei K. 5-Hydroxymethylfurfural Mitigates Lipopolysaccharide-Stimulated Inflammation via Suppression of MAPK, NF-κB and mTOR Activation in RAW 264.7 Cells. Molecules 2019; 24:molecules24020275. [PMID: 30642099 PMCID: PMC6359491 DOI: 10.3390/molecules24020275] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 01/01/2023] Open
Abstract
5-Hydroxymethylfurfural (5-HMF) is found in many food products including honey, dried fruits, coffee and black garlic extracts. Here, we investigated the anti-inflammatory activity of 5-HMF and its underlying mechanisms in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. 5-HMF pretreatment ranging from 31.5 to 126.0 μg/mL reduced the production of nitric oxide (NO), prostaglandin E2 (PGE2) and pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) in a concentration-dependent manner in LPS-stimulated cells. Moreover, 5-HMF-pretreated cells significantly down-regulated the mRNA expression of two major inflammatory mediators, nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) and suppressed the production of pro-inflammatory cytokines, as compared with the only LPS-stimulated cells. 5-HMF suppressed the phosphorylation of extracellular regulated protein kinases (ERK1/2), c-Jun N-terminal kinase (JNK), IκBα, NF-κB p65, the mammalian target of rapamycin (mTOR) and protein kinase B (Akt). Besides, 5-HMF was proved to inhibit NF-κB p65 translocation into nucleus to activate inflammatory gene transcription. These results suggest that 5-HMF could exert the anti-inflammatory activity in the LPS-induced inflammatory response by inhibiting the MAPK, NF-κB and Akt/mTOR pathways. Thus, 5-HMF could be considered as a therapeutic ingredient in functional foods.
Collapse
Affiliation(s)
- Fanhui Kong
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, Guangdong, China.
| | - Bae Hoon Lee
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou 325011, Zhejiang, China.
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.
| | - Kun Wei
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, Guangdong, China.
- Wenzhou Institute of Biomaterials and Engineering, CAS, Wenzhou 325011, Zhejiang, China.
| |
Collapse
|
199
|
Total flavonoids extracted from Nervilia Fordii function in polycystic ovary syndrome through IL-6 mediated JAK2/STAT3 signaling pathway. Biosci Rep 2019; 39:BSR20181380. [PMID: 30463907 PMCID: PMC6328881 DOI: 10.1042/bsr20181380] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/01/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Abstract
Large doses of flavonoids could cure many diseases with no serious side effects. However, the role of flavonoids in the treatment of polycystic ovary syndrome (PCOS) has not been reported. Therefore, total flavonoids extracted from Nervilia Fordii were selected to explore its therapeutic efficiency in PCOS. PCOS rat model was constructed to explore the role of total flavonoids in the treatment of PCOS. ELISA was used to assess the changes of ovulation function under the treatment of total flavonoids with or without exogenous interleukin-6 (IL-6). Western blot, real-time PCR and immunohistochemistry were carried out to assess the related molecular mechanisms. We explored that total flavonoids obviously increased the serum levels of follicle-stimulating hormone (FSH), and sharply decreased the serum levels of luteinizing hormone (LH), testosterone (T) and insulin (INS) in the PCOS-IR rats via partly inhibiting the activation of JAK2/STAT3 pathway, partially up-regulating the IL-6 expression and partially down-regulating the suppressor of cytokine signaling 3 (SOCS3) expression in ovaries of PCOS rats. The effect of total flavonoids on estrous cycles, serum levels of FSH, LH, T and INS were partially attenuated by IL-6 in PCOS rat model. Moreover, IL-6 significantly reversed the effect of total flavonoids on the phosphorylation of JAK2/STAT3, the expression of IL-6 and SOCS3 in ovaries of PCOS rats. Total flavonoids extracted from Nervilia Fordii might induce the expression of IL-6 in ovary and act as a potential therapeutic drug for the treatment of PCOS.
Collapse
|
200
|
Nutrition and Cardiovascular Health. Int J Mol Sci 2018; 19:ijms19123988. [PMID: 30544955 PMCID: PMC6320919 DOI: 10.3390/ijms19123988] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 11/30/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in Western countries, representing almost 30% of all deaths worldwide. Evidence shows the effectiveness of healthy dietary patterns and lifestyles for the prevention of CVD. Furthermore, the rising incidence of CVD over the last 25 years has become a public health priority, especially the prevention of CVD (or cardiovascular events) through lifestyle interventions. Current scientific evidence shows that Western dietary patterns compared to healthier dietary patterns, such as the ‘Mediterranean diet’ (MeDiet), leads to an excessive production of proinflammatory cytokines associated with a reduced synthesis of anti-inflammatory cytokines. In fact, dietary intervention allows better combination of multiple foods and nutrients. Therefore, a healthy dietary pattern shows a greater magnitude of beneficial effects than the potential effects of a single nutrient supplementation. This review aims to identify potential targets (food patterns, single foods, or individual nutrients) for preventing CVD and quantifies the magnitude of the beneficial effects observed. On the other hand, we analyze the possible mechanisms implicated in this cardioprotective effect.
Collapse
|