1
|
Gholami Z, Maracy MR, Paknahad Z. The effects of MIND diet and propolis supplementation on metabolic syndrome: A randomized controlled clinical trial. Heliyon 2024; 10:e34493. [PMID: 39220895 PMCID: PMC11363718 DOI: 10.1016/j.heliyon.2024.e34493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
The MIND is a novel eating plan preserves cognitive function. Propolis is a resinous substance that has several biological and medicinal properties. This study examines the effect of the MIND diet and propolis supplementation on MetS indices among metabolic syndrome subjects. This RCT study, was conducted on adults with metabolic syndrome who were referred to the Hazrat Ali Health Center in Isfahan. 84 eligible subjects were divided into 3 groups. Including MIND diet + Propolis supplement, MIND diet + placebo, and control group. The data obtained from the subjects was analyzed in two descriptive and analytic levels. The Shapiro-Wilk test and examination of skewness were conducted to assess the normality of the distribution of quantitative variables. Quantitative variables were reported using either the mean (SD). SPSS Statistics software version 26 was used for statistical analysis of data. In this study the MIND + Propolis group compared to the control group after adjusting variables showed a significant decrease (p-value < 0.05) in weight, BMI, WC, SBP, DBP, and TG by 0.97 times (3%), 0.97 times (3%), 0.98 times (2%), 0.93 times (7%), 0.94 times (6%), and 0.75 times (25%), respectively; this significant change was also observed in FBS (p-value < 0.001) by 0.85 times (15%), and HDL-C (mg/dl) has shown a significant increase (p-value < 0.05) by 1.17 times (17%). MIND group compared to the control group after adjusting variables showed a significant decrease (p-value < 0.05) in BMI, WC, and SBP by 0.98 times (2%), 0.98 times (2%), and 0.95 times (5%), respectively; this significant change (p-value < 0.001) was also observed in DBP, FBS, and TG by 0.92 times (8%), 0.83 times (17%), and 0.71 times (29%), respectively; HDL-C has shown a significant increase (p-value < 0.001) by 1.21 times (21%), and weight has shown a non-significant decrease (p-value = 0.055) by 0.98 times (2%). This study indicated that the MIND diet + Propolis supplement and MIND diet compared to the control group can significantly decrease BMI, WC, SBP, DBP, FBS, TG, and weight (non-significant for the MIND group), and also increase HDL-C.
Collapse
Affiliation(s)
- Zainab Gholami
- School of Nutrition and Food Science, Students' Research Committee, Department of Clinical Nutrition, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Reza Maracy
- Department of Epidemiology and Biostatistics, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zamzam Paknahad
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
2
|
Gholami Z, Maracy MR, Paknahad Z. Effects of MIND diet and propolis supplementation on metabolic syndrome indices and cognitive function among patients with metabolic syndrome in Isfahan, Iran, 2024. A rationale and study protocol for randomized controlled trial. Health Sci Rep 2024; 7:e2262. [PMID: 39135701 PMCID: PMC11318029 DOI: 10.1002/hsr2.2262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/22/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Metabolic syndrome (MetS) is the main general and clinical health challenge worldwide. Based on the National Cholesterol Education Program, if the person has three or more indices containing: elevated fasting blood sugar, high levels of triglycerides, hypertension, low levels of high-density lipoprotein cholesterol, and central obesity, he suffers MetS. The Mediterranean-Dietary Approaches to Stop Hypertension (DASH) Intervention for Neurodegenerative Delay diet is a novel diet that with the specific aim of safeguarding cognitive function. Propolis is a resinous substance produced by bees from the combination of buds and secretions of plants with saliva and bee enzymes. After propolis supplementation, a significant reduction in fasting plasma glucose levels and lipid profiles has been observed. Considering the importance of chronic diseases like MetS on health, the role of the Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diet and propolis supplement that will improve blood sugar, blood lipid, anthropometric indicators, blood pressure, and cognitive function, and limited contradictory studies, we decided to conduct this study. This study, which is a randomized controlled clinical trial study, will be conducted on adults with MetS who will visit Hazrat Ali Health Center in Isfahan. Participants must provide informed consent before engaging in the study. Demographic data such as age, gender, and medical history will be recorded. Then, anthropometric indices, MetS indices, and cognitive function will be measured in all subjects. The study participants will be divided into three groups and will be controlled for 12 weeks. We will have a MIND diet + placebo group, a MIND diet + propolis supplement group, and a control group that will receive a microcrystalline cellulose placebo and usual dietary advice. At the end of the intervention, all indices will be assessed again. The data obtained in the study will be analyzed at descriptive and analytical levels by the statistical software SPSS26. The present study's protocol was approved by the Iranian Registry of Clinical Trials (www.irct.ir) on 3/28/2023 and a registration reference is IRCT20230105057054N1.
Collapse
Affiliation(s)
- Zeinab Gholami
- Department of Clinical Nutrition, School of Nutrition and Food Science, Students' Research CommitteeIsfahan University of Medical SciencesIsfahanIran
| | - Mohammad Reza Maracy
- Department of Epidemiology and Biostatistics, School of HealthIsfahan University of Medical SciencesIsfahanIran
| | - Zamzam Paknahad
- Department of Clinical Nutrition, Faculty of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
3
|
Gravesteijn E, Adam JJ, Mensink RP, Winkens B, Plat J. Effects of the egg protein hydrolysate NWT-03 on cognitive function in men and women with the metabolic syndrome: a randomized, double-blind, placebo-controlled study. Nutr Neurosci 2023; 26:1212-1221. [PMID: 36373820 DOI: 10.1080/1028415x.2022.2144204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives: The metabolic syndrome is associated with cardiovascular diseases and cognitive decline. The egg protein hydrolysate NWT-03 has shown to improve cardiovascular risk factors in humans. This study investigated whether NWT-03 also has an effect on cognitive function.Methods: Men and women with the metabolic syndrome (n = 76) with a mean age of 60 ± 10 years participated in this randomized, double-blind, placebo-controlled, cross-over trial with an intervention (5 g/day NWT-03) and control period (5 g/day maltodextrin) of 4 weeks separated by a wash-out period of 2-8 weeks. Cognitive function was assessed with the anti-cue reaction time test (impulse control) and psychomotor vigilance test (sustained attention) at day 0, 2, and 27 of both periods. Serum brain-derived neurotrophic factor (BDNF) concentrations were measured at the start and end of both periods.Results: NWT-03 consumption significantly improved the change (day 27 - day 0) in response times of the anti-cue reaction time test compared with the control period (P < 0.001), but not of the psychomotor vigilance test (P = 0.487). Serum BDNF concentrations of all subjects did not significantly change (P = 0.241).Conclusion: NWT-03 has the ability to improve cognitive function within the executive function domain. The underlying mechanism warrants further research and could either be indirect via inhibition of dipeptidyl peptidase 4 (DPP4) or direct via passage of small peptides over the blood-brain barrier inducing local effects.Trial registration: ClinicalTrials.gov identifier: NCT02561663.
Collapse
Affiliation(s)
- Elske Gravesteijn
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+ (MUMC+), Maastricht, Netherlands
| | - Jos J Adam
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+ (MUMC+), Maastricht, Netherlands
| | - Ronald P Mensink
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+ (MUMC+), Maastricht, Netherlands
| | - Bjorn Winkens
- Department of Methodology and Statistics, CAPHRI School for Public Health and Primary Care, Maastricht University Medical Center+ (MUMC+), Maastricht, Netherlands
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+ (MUMC+), Maastricht, Netherlands
| |
Collapse
|
4
|
Wang XF, Chen X, Tang Y, Wu JM, Qin DL, Yu L, Yu CL, Zhou XG, Wu AG. The Therapeutic Potential of Plant Polysaccharides in Metabolic Diseases. Pharmaceuticals (Basel) 2022; 15:1329. [PMID: 36355500 PMCID: PMC9695998 DOI: 10.3390/ph15111329] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/15/2022] [Accepted: 10/25/2022] [Indexed: 07/29/2023] Open
Abstract
Plant polysaccharides (PPS) composed of more than 10 monosaccharides show high safety and various pharmacological activities, including immunoregulatory, antitumor, antioxidative, antiaging, and other effects. In recent years, emerging evidence has indicated that many PPS are beneficial for metabolic diseases, such as cardiovascular disease (CVD), diabetes, obesity, and neurological diseases, which are usually caused by the metabolic disorder of fat, sugar, and protein. In this review, we introduce the common characteristics and functional activity of many representative PPS, emphasize the common risks and molecular mechanism of metabolic diseases, and discuss the pharmacological activity and mechanism of action of representative PPS obtained from plants including Aloe vera, Angelica sinensis, pumpkin, Lycium barbarum, Ginseng, Schisandra chinensis, Dioscorea pposite, Poria cocos, and tea in metabolic diseases. Finally, this review will provide directions and a reference for future research and for the development of PPS into potential drugs for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Xiao-Fang Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| | - Xue Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, College of Pharmacy, Changsha Medical University, Changsha 410219, China
| |
Collapse
|
5
|
Ruyvaran M, Zamani A, Mohamadian A, Zarshenas MM, Eftekhari MH, Pourahmad S, Abarghooei EF, Akbari A, Nimrouzi M. Safflower (Carthamus tinctorius L.) oil could improve abdominal obesity, blood pressure, and insulin resistance in patients with metabolic syndrome: A randomized, double-blind, placebo-controlled clinical trial. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114590. [PMID: 34487844 DOI: 10.1016/j.jep.2021.114590] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/19/2021] [Accepted: 08/29/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Carthamus tinctorius L. (Safflower) has been widely recommended to treat metabolic disorders in traditional herbal medicine in Persia, China, Korea, Japan, and other East-Asian countries. The anti-hypercholesterolemic and antioxidant effects of this plant have been well documented, but its protective effects against Metabolic Syndrome (MetS) have not been fully illustrated. AIM OF THE STUDY The present study aimed to evaluate the effects of safflower oil on MetS risk factors. MATERIALS AND METHODS In this randomized, double-blind, placebo-controlled clinical trial, 67 patients with MetS were administered either divided 8 g safflower oil or placebo daily for 12 weeks. All patients were advised to follow their previous diets and physical activities. RESULTS Safflower oil resulted in a significant reduction in waist circumference (-2.42 ± 3.24 vs. 0.97 ± 2.53, p<0.001), systolic blood pressure (-8.80 ± 9.77 vs. -2.26 ± 8.56, p = 0.021), diastolic blood pressure (-3.53 ± 7.52 vs. -0.70 ± 6.21, p = 0.041), fasting blood sugar (-5.03 ± 10.62 vs. 2.94 ± 7.57, p = 0.003), and insulin resistance (-0.59 ± 1.43 vs. 0.50 ± 1, p = 0.012), but an increase in adiponectin level (0.38 ± 0.99 vs. -0.09 ± 0.81, p = 0.042) in the treatment group in comparison to the placebo group. The results revealed a direct relationship between leptin level and Body Mass Index (BMI) in both groups (p<0.001). In addition, increase in BMI resulted in a non-significant decrease in adiponectin level in both groups. Moreover, no significant difference was observed between the two groups regarding lipid profiles, leptin serum level, serum creatinine concentration, and other outcomes. CONCLUSION Safflower oil without lifestyle modification improved abdominal obesity, blood pressure, and insulin resistance in patients with MetS.
Collapse
Affiliation(s)
- Maede Ruyvaran
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Internal Medicine, Endocrine and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Traditional Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Zamani
- Department of Internal Medicine, Endocrine and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Alireza Mohamadian
- Department of Radiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad M Zarshenas
- Medicinal Plants Processing Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Phytopharmaceuticals (Traditional Pharmacy), School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Hassan Eftekhari
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Saeedeh Pourahmad
- Department of Biostatics, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ebrahim Fallahzadeh Abarghooei
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Traditional Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Abolfazl Akbari
- Department of Physiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Majid Nimrouzi
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Traditional Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
6
|
Effectiveness of a complex psychosocial intervention to reduce metabolic syndrome in psychiatric outpatients with severe/persistent mental illness. CURRENT PSYCHOLOGY 2021; 42:9915-9924. [PMID: 34539154 PMCID: PMC8435195 DOI: 10.1007/s12144-021-02269-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2021] [Indexed: 11/21/2022]
Abstract
The prevalence of metabolic syndrome among individuals with severe mental illness is considerably higher than in the general population, contributing to the 15–20-year shorter life expectancy of this client population. The aim of this pilot study was to evaluate the effectiveness of a novel, complex psychosocial program to reduce metabolic syndrome. Members of both the intervention (n = 78) and control (n = 31) group were psychiatric outpatients with severe/persistent mental illness struggling with one or more symptoms of metabolic syndrome. Beyond the default elements of similar programs such as diet and exercise, the intervention covered medication use, sleep hygiene, stress management, as well as addressing spiritual needs, mindfulness, addictions, and self-care. Assessment of metabolic indicators were completed at baseline, at the end of the 11-week intervention, and 6 months post-intervention. The trajectory of change over time was significantly more favorable in the treatment than in the control group in terms of waist circumference (p = 0.013, η2 = 0.093) and a positive trend emerged in relation to blood glucose level (p = 0.082, η2 = 0.057). However, no statistically reliable difference was observed between the intervention and the control group regarding the other outcome variables (body mass index, systolic and diastolic blood pressure, serum triglyceride level, serum HDL cholesterol level, overall metabolic syndrome severity). These findings suggest that to produce more robust benefits, psychosocial interventions targeting the metabolic health of individuals with complex mental health needs should be either longer in duration if resources permit or narrower in focus (diet and exercise mainly) if resources are scarce.
Collapse
|
7
|
Jo A, Lee Y, Kam TI, Kang SU, Neifert S, Karuppagounder SS, Khang R, Kang H, Park H, Chou SC, Oh S, Jiang H, Swing DA, Ham S, Pirooznia S, Umanah GKE, Mao X, Kumar M, Ko HS, Kang HC, Lee BD, Lee YI, Andrabi SA, Park CH, Lee JY, Kim H, Kim H, Kim H, Cho JW, Paek SH, Na CH, Tessarollo L, Dawson VL, Dawson TM, Shin JH. PARIS farnesylation prevents neurodegeneration in models of Parkinson's disease. Sci Transl Med 2021; 13:13/604/eaax8891. [PMID: 34321320 PMCID: PMC9990146 DOI: 10.1126/scitranslmed.aax8891] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 12/09/2020] [Accepted: 05/20/2021] [Indexed: 12/18/2022]
Abstract
Accumulation of the parkin-interacting substrate (PARIS; ZNF746), due to inactivation of parkin, contributes to Parkinson's disease (PD) through repression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α; PPARGC1A) activity. Here, we identify farnesol as an inhibitor of PARIS. Farnesol promoted the farnesylation of PARIS, preventing its repression of PGC-1α via decreasing PARIS occupancy on the PPARGC1A promoter. Farnesol prevented dopaminergic neuronal loss and behavioral deficits via farnesylation of PARIS in PARIS transgenic mice, ventral midbrain transduction of AAV-PARIS, adult conditional parkin KO mice, and the α-synuclein preformed fibril model of sporadic PD. PARIS farnesylation is decreased in the substantia nigra of patients with PD, suggesting that reduced farnesylation of PARIS may play a role in PD. Thus, farnesol may be beneficial in the treatment of PD by enhancing the farnesylation of PARIS and restoring PGC-1α activity.
Collapse
Affiliation(s)
- Areum Jo
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea.,Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yunjong Lee
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea.,Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Stewart Neifert
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Senthilkumar S Karuppagounder
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Rin Khang
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea
| | - Hojin Kang
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea.,Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyejin Park
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shih-Ching Chou
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sungtaek Oh
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Haisong Jiang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Deborah A Swing
- Neural Development Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21705, USA
| | - Sangwoo Ham
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea
| | - Sheila Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - George K E Umanah
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Manoj Kumar
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Han Seok Ko
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Ho Chul Kang
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Byoung Dae Lee
- Neurodegeneration Control Research Center, Department of Neuroscience, Department of Physiology, Kyung Hee University School of Medicine, Seoul 02447, South Korea
| | - Yun-Il Lee
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shaida A Andrabi
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chi-Hu Park
- Research Core Facility, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea
| | - Ji-Yeong Lee
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea
| | - Hanna Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea
| | - Hyein Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea.,Research Core Facility, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea
| | - Hyojung Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea
| | - Jin Whan Cho
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Chan Hyun Na
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lino Tessarollo
- Neural Development Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21705, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130-2685, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joo-Ho Shin
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, South Korea. .,Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Samsung Medical Center, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Seoul 06351, South Korea
| |
Collapse
|
8
|
Xavier-Santos D, Bedani R, Lima ED, Saad SMI. Impact of probiotics and prebiotics targeting metabolic syndrome. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103666] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
9
|
Puente D, López-Jiménez T, Cos-Claramunt X, Ortega Y, Duarte-Salles T. Metabolic syndrome and risk of cancer: a study protocol of case-control study using data from the Information System for the Development of Research in Primary Care (SIDIAP) in Catalonia. BMJ Open 2019; 9:e025365. [PMID: 31201184 PMCID: PMC6575640 DOI: 10.1136/bmjopen-2018-025365] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Metabolic syndrome (MS) is defined by the clustering of specific metabolic disorders in one subject. MS is highly prevalent globally and currently considered a growing public health concern. MS comprises obesity, hypertension, dyslipidaemia and insulin resistance. Mechanisms linking MS with cancer are poorly understood, and it is as yet unknown if MS confers a greater risk than the risk entailed by each of its separate components. The main objective of this study is to compare the association between MS and 14 site-specific cancer against the association between one or two individual components of MS and cancer. The secondary objective is to evaluate the time elapsed since the diagnosis of MS and the subsequent onset of cancer within the 2006-2017 period by sex. METHODS AND ANALYSIS A case-control study will be conducted for the main objective and a cohort of patients with MS will be followed for the evaluation of the second objective. Incident cases of fourteen types of cancer in patients ≥40 years of age diagnosed prospectively will be selected from electronic primary care records in the Information System for Research in Primary Care (SIDIAP database; www.sidiap.org). The SIDIAP database includes anonymous data from 6 million people (80% of the Catalan population) registered in 286 primary healthcare centres. Each matched control (four controls for each case) will have the same inclusion date, the same sex and age (±1 year) than the paired case. Logistic regression and a descriptive analysis and Kaplan-Meier analysis will be performed, in accordance with the objectives. ETHICS AND DISSEMINATION The protocol of the study was approved by the IDIAP Jordi Gol Clinical Research Ethics Committee (protocol P17/212). The study's findings will be published in a peer-reviewed journal and disseminated at national and international conferences and oral presentations to researchers, clinicians and policy makers.
Collapse
Affiliation(s)
- Diana Puente
- Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Tomàs López-Jiménez
- Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Xavier Cos-Claramunt
- Foundation Network of Study Groups of Diabetes in Primary Care (redGDPS), Sabadell, Spain
- Primary Care Centre Sant Martí de Provençals. Primary Care Management Barcelona Ciutat, Catalan Institute of Health, Barcelona, Spain
| | - Yolanda Ortega
- CAP Salou, Institut Català de la Salut, Tarragona, Spain
| | - Talita Duarte-Salles
- Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Barcelona, Spain
| |
Collapse
|
10
|
Bellastella G, Scappaticcio L, Esposito K, Giugliano D, Maiorino MI. Metabolic syndrome and cancer: "The common soil hypothesis". Diabetes Res Clin Pract 2018; 143:389-397. [PMID: 29807099 DOI: 10.1016/j.diabres.2018.05.024] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
Abstract
Metabolic syndrome (MetS) and cancer share many modifiable risk factors including age, genetic factors, obesity, physical inactivity, unhealthy diet, alcohol, smoking, endocrine disruptors exposure, circadian clock disturbances, and air pollution. MetS is closely linked to cancer, as it increases cancer risk and cancer-related mortality; moreover, cancer survivors have an increased risk of MetS. Elucidating the mechanisms linking MetS to cancer is important to prevent or delay these two conditions. Possible mechanisms explaining the relationship between MetS and cancer include hyperinsulinemia and alterations of insulin-like growth factor system, chronic subclinical inflammation, abnormalities in sex hormones metabolism and adipokines, hyperglicemia, alterations in both gene expression and hormonal profile by endocrine disruptors and air pollution, desynchronization of circadian clock. The common soil hypothesis claims that MetS may be considered a surrogate marker for dietary risk factors of cancer, and a warning sign for susceptible individuals exposed to an unhealthy diet. The common soil hypothesis and the clepsydra of foods represent a theoretical substrate to preventive intervention strategies against the pandemics of MetS and cancer: adherence to healthy dietary patterns is associated with a reduced risk of MetS, and improvement of the quality of diet is consistently associated with a reduction in cancer-related mortality.
Collapse
Affiliation(s)
- Giuseppe Bellastella
- Unit of Endocrinology and Metabolic Diseases, Department of Medical, Surgical, Neurological, Metabolic Sciences and Aging, University of Campania "L. Vanvitelli", Piazza L. Miraglia n° 2, 80138 Naples, Italy.
| | - Lorenzo Scappaticcio
- Unit of Endocrinology and Metabolic Diseases, Department of Medical, Surgical, Neurological, Metabolic Sciences and Aging, University of Campania "L. Vanvitelli", Piazza L. Miraglia n° 2, 80138 Naples, Italy.
| | - Katherine Esposito
- Diabetes Unit, Department of Medical, Surgical, Neurological, Metabolic Sciences and Aging, University of Campania "L. Vanvitelli", Piazza L. Miraglia n° 2, 80138 Naples, Italy; Associazione Salute con Stile, Piazza L. Miraglia n° 2, 80138 Naples, Italy.
| | - Dario Giugliano
- Unit of Endocrinology and Metabolic Diseases, Department of Medical, Surgical, Neurological, Metabolic Sciences and Aging, University of Campania "L. Vanvitelli", Piazza L. Miraglia n° 2, 80138 Naples, Italy; Associazione Salute con Stile, Piazza L. Miraglia n° 2, 80138 Naples, Italy.
| | - Maria Ida Maiorino
- Unit of Endocrinology and Metabolic Diseases, Department of Medical, Surgical, Neurological, Metabolic Sciences and Aging, University of Campania "L. Vanvitelli", Piazza L. Miraglia n° 2, 80138 Naples, Italy.
| |
Collapse
|
11
|
Jung TW, Chung YH, Kim HC, Abd El-Aty AM, Jeong JH. Hyperlipidemia-induced hepassocin in the liver contributes to insulin resistance in skeletal muscle. Mol Cell Endocrinol 2018; 470:26-33. [PMID: 29111387 DOI: 10.1016/j.mce.2017.10.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022]
Abstract
Hepassocin (HPS) has recently been identified as a novel hepatokine that causes hepatic steatosis. However, the role of HPS in the development of insulin resistance in skeletal muscle under obesity remains unclear. The effect of hyperlipidemia on hepatic HPS expression was evaluated in primary hepatocytes and liver of mice. HPS-mediated signal pathways were explored using small interfering (si) RNAs of specific genes or inhibitors. We found that treatment of primary hepatocytes with palmitate could induce HPS expression through C/EBPβ-mediated transcriptional activation. Furthermore, increased HPS expression was observed in the liver of high fat diet (HFD)-fed or tunicamycin-treated mice. Pretreatment with 4-phenylbutyrate (4-BPA) (an endoplasmic reticulum (ER) stress inhibitor) and suppression of p38 by siRNA abrogated the effect of palmitate on HPS expression in primary hepatocytes. Treatment of differentiated C2C12 cells with recombinant HPS caused c-Jun N-terminal kinase (JNK) phosphorylation and impairment of insulin sensitivity in a dose-dependent manner. siRNA-mediated suppression of JNK reduced the effect of HPS on insulin signaling. Furthermore, the suppression of epidermal growth factor receptor (EGFR) by siRNA mitigated both HPS-induced JNK phosphorylation and insulin resistance. In addition, HPS did not affect inflammation and ER stress in differentiated C2C12 cells. In conclusion, we elucidated that ER stress induced by palmitate could increase the expression of HPS in hepatocytes and further contribute to the development of insulin resistance in skeletal muscle via EGFR/JNK-mediated pathway. Taken together, we suggest that HPS could be a therapeutic target for obesity-linked insulin resistance.
Collapse
Affiliation(s)
- Tae Woo Jung
- Research Administration Team, Seoul National University Bundang Hospital, Gyeonggi, Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt; Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Republic of Korea; Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum 25240, Turkey.
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Current perspectives between metabolic syndrome and cancer. Oncotarget 2018; 7:38959-38972. [PMID: 27029038 PMCID: PMC5122443 DOI: 10.18632/oncotarget.8341] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/20/2016] [Indexed: 12/21/2022] Open
Abstract
Metabolic syndrome is a cluster of risk factors that lead to cardiovascular morbidity and mortality. Recent studies linked metabolic syndrome and several types of cancer. Although metabolic syndrome may not necessarily cause cancer, it is linked to poorer cancer outcomes including increased risk of recurrence and overall mortality. This review tends to discuss the major biological and physiological alterations involved in the increase of incidence and mortality of cancer patients affected by metabolic syndrome. We focus on metabolic syndrome-associated visceral adiposity, hyperinsulinemia, hyperglycemia, insulin-like growth factor (IGF-I) pathway as well as estrogen signaling and inflammation. Several of these factors are also involved in carcinogenesis and cancer progression. A better understanding of the link between metabolic syndrome and cancer may provide new insight about oncogenesis. Moreover, prevention of metabolic syndrome - related alterations may be an important aspect in the management of cancer patients during simultaneous palliative care.
Collapse
|
13
|
Abrat OB, Storey JM, Storey KB, Lushchak VI. High amylose starch consumption induces obesity in Drosophila melanogaster and metformin partially prevents accumulation of storage lipids and shortens lifespan of the insects. Comp Biochem Physiol A Mol Integr Physiol 2018; 215:55-62. [DOI: 10.1016/j.cbpa.2017.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 11/30/2022]
|
14
|
Takahama U, Hirota S. Interactions of flavonoids with α-amylase and starch slowing down its digestion. Food Funct 2018; 9:677-687. [DOI: 10.1039/c7fo01539a] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hydrophobic flavonoids can suppress starch digestion in the intestine by forming starch-flavonoid complexes.
Collapse
Affiliation(s)
- Umeo Takahama
- Department of Health and Nutritional Care
- Faculty of Allied Health Sciences
- University of East Asia
- Shimonoseki
- Japan
| | - Sachiko Hirota
- Department of Health and Nutritional Care
- Faculty of Allied Health Sciences
- University of East Asia
- Shimonoseki
- Japan
| |
Collapse
|
15
|
Robberecht H, De Bruyne T, Hermans N. Effect of various diets on biomarkers of the metabolic syndrome. Int J Food Sci Nutr 2016; 68:627-641. [PMID: 28027691 DOI: 10.1080/09637486.2016.1269726] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The impact of the normal whole diet in different countries and of special types of diet on the biomarkers of the metabolic syndrome (MetS) is reviewed. Diet type, specification, risk of MetS and studied biomarkers, as far as could be traced, are included. Critical points in published studies are mentioned. Description of the traditional dietary patterns for the various countries is not always well-defined and numbers of persons in the studied population are sometimes quite limited, which hamper drawing definite conclusions. Since a Nordic diet, a dietary approach to stop hypertension (DASH), and especially a Mediterranean diet are quite promising, due to its health claims, the food pattern is studied more in detail and a large spectrum of vegetarian diets are included as well. Most of the time lipid profile and high sensitive C-reactive protein (hsCRP) are the studied biomarkers in response to diet intake.
Collapse
Affiliation(s)
- Harry Robberecht
- a Department of Pharmaceutical Sciences, Laboratory of Nutrition and Functional Foods , NatuRA (Natural Products and Food-Research and Analysis), University of Antwerp , Antwerp , Belgium
| | - Tess De Bruyne
- a Department of Pharmaceutical Sciences, Laboratory of Nutrition and Functional Foods , NatuRA (Natural Products and Food-Research and Analysis), University of Antwerp , Antwerp , Belgium
| | - Nina Hermans
- a Department of Pharmaceutical Sciences, Laboratory of Nutrition and Functional Foods , NatuRA (Natural Products and Food-Research and Analysis), University of Antwerp , Antwerp , Belgium
| |
Collapse
|
16
|
Westerink NL, Nuver J, Lefrandt JD, Vrieling AH, Gietema JA, Walenkamp AME. Cancer treatment induced metabolic syndrome: Improving outcome with lifestyle. Crit Rev Oncol Hematol 2016; 108:128-136. [PMID: 27931830 DOI: 10.1016/j.critrevonc.2016.10.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 08/26/2016] [Accepted: 10/26/2016] [Indexed: 12/19/2022] Open
Abstract
Increasing numbers of long-term cancer survivors face important treatment related adverse effects. Cancer treatment induced metabolic syndrome (CTIMetS) is an especially prevalent and harmful condition. The aetiology of CTIMetS likely differs from metabolic syndrome in the general population, but effective treatment and prevention methods are probably similar. In this review, we summarize the potential mechanisms leading to the development of CTIMetS after various types of cancer treatment. Furthermore, we propose a safe and accessible method to treat or prevent CTIMetS through lifestyle change. In particular, we suggest that a lifestyle intervention and optimization of energy balance can prevent or mitigate the development of CTIMetS, which may contribute to optimal survivorship care.
Collapse
Affiliation(s)
- N L Westerink
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - J Nuver
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - J D Lefrandt
- Department of Vascular Medicine, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - A H Vrieling
- Department of Rehabilitation Medicine, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - J A Gietema
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - A M E Walenkamp
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
17
|
Kouvari M, Notara V, Kalogeropoulos N, Panagiotakos DB. Diabetes mellitus associated with processed and unprocessed red meat: an overview. Int J Food Sci Nutr 2016; 67:735-43. [PMID: 27309597 DOI: 10.1080/09637486.2016.1197187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
According to American Diabetes Association "as many as 1 in 3 American adults will have diabetes by 2050" imposing a serious burden on healthcare services and highlighting a substantial need to reduce "new-cases" incidence. Diabetes is inextricably linked to diet, in the prevention-spectrum. Red-meat-intake has been positively associated with reduced glycemic control. However, divergence exists among meat subtypes (i.e. fresh and processed) and the magnitude of their impact on diabetes development. The present overview attempted to summarize the latest data regarding red-meat subtypes on the examined association. Four meta-analysis and 10 prospective studies, focusing on the role of fresh and processed red meat in diabetes prevention, were selected. All of studies highlighted the aggravating role of processed meat-products in diabetes incidence, while fresh meat reached significance in only half of them. Therefore, the contribution of fresh red meat on diabetes remains inconclusive. Valid conclusions seem more robust concerning processed-meat-consumption.
Collapse
Affiliation(s)
- M Kouvari
- a Department of Nutrition - Dietetics , School of Health Science and Education, Harokopio University , Athens , Greece
| | - V Notara
- a Department of Nutrition - Dietetics , School of Health Science and Education, Harokopio University , Athens , Greece
| | - N Kalogeropoulos
- a Department of Nutrition - Dietetics , School of Health Science and Education, Harokopio University , Athens , Greece
| | - D B Panagiotakos
- a Department of Nutrition - Dietetics , School of Health Science and Education, Harokopio University , Athens , Greece
| |
Collapse
|
18
|
Rajabi S, Mazloom Z, Zamani A, Tabatabaee HR. Effect of Low Glycemic Index Diet Versus Metformin on Metabolic Syndrome. Int J Endocrinol Metab 2015; 13:e23091. [PMID: 26587028 PMCID: PMC4648126 DOI: 10.5812/ijem.23091] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 03/18/2015] [Accepted: 06/16/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) continues to be highly prevalent and contributes to a rapidly growing problem worldwide. The most important therapeutic intervention for metabolic syndrome is diet modification, an intervention whose efficacy has been proven for metabolic syndrome. OBJECTIVES The aim of the present study was to compare the effects of low glycemic index diet versus metformin on MetS components in adults with MetS. PATIENTS AND METHODS Fifty-one adults with MetS participated in this randomized controlled clinical trial. Patients were randomly allocated to two groups of metformin and low glycemic index diet. The intervention period was eight weeks. The studied participants were compared at baseline and the end of the trial, regarding the following factors: weight, blood pressure, waist circumference, fasting blood sugar, hemoglobin A1c and lipid profiles (Triglyceride (TG), Total Cholesterol (TC), Low-Density Lipoprotein (LDL) cholesterol, and High-Density Lipoprotein (HDL) cholesterol). RESULTS The anthropometric measurements, Fasting Blood Sugar (FBS), Hemoglobin A1c, serum lipid profiles (TG, TC, LDL-C, HDL-C) and lipoprotein ratio (LDL/HDL) showed a significant decrease after the intervention in both groups (P < 0.05). Comparison of the difference between the two groups was not significant, except for the mean reduction in FBS, which was more in the metformin group although this was not clinically significant. CONCLUSIONS This study supports the assumption that low glycemic index diet as well as metformin can positively affect metabolic syndrome components.
Collapse
Affiliation(s)
- Shirin Rajabi
- Student Research Committee, Department of Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Zohreh Mazloom
- Department of Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, IR Iran
- Corresponding author: Zohreh Mazloom, Department of Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, IR Iran. Tel: +98-7137251001, Fax. +98-7137260225, E-mail:
| | - Ali Zamani
- Endocrine and Metabolism Research Center, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Hamid Reza Tabatabaee
- Department of Epidemiology, Research Center for Health Sciences, School of Health , Shiraz University of Medical Sciences, Shiraz, IR Iran
| |
Collapse
|
19
|
Sottero B, Gargiulo S, Russo I, Barale C, Poli G, Cavalot F. Postprandial Dysmetabolism and Oxidative Stress in Type 2 Diabetes: Pathogenetic Mechanisms and Therapeutic Strategies. Med Res Rev 2015; 35:968-1031. [PMID: 25943420 DOI: 10.1002/med.21349] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Postprandial dysmetabolism in type 2 diabetes (T2D) is known to impact the progression and evolution of this complex disease process. However, the underlying pathogenetic mechanisms still require full elucidation to provide guidance for disease prevention and treatment. This review focuses on the marked redox changes and inflammatory stimuli provoked by the spike in blood glucose and lipids in T2D individuals after meals. All the causes of exacerbated postprandial oxidative stress in T2D were analyzed, also considering the consequence of enhanced inflammation on vascular damage. Based on this in-depth analysis, current strategies of prevention and pharmacologic management of T2D were critically reexamined with particular emphasis on their potential redox-related rationale.
Collapse
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Turin, 10043, Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Turin, 10043, Italy
| | - Isabella Russo
- Internal Medicine and Metabolic Disease Unit, Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Turin, 10043, Italy
| | - Cristina Barale
- Internal Medicine and Metabolic Disease Unit, Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Turin, 10043, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Turin, 10043, Italy
| | - Franco Cavalot
- Internal Medicine and Metabolic Disease Unit, Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Turin, 10043, Italy
| |
Collapse
|
20
|
Wong YV, Cook P, Somani BK. The association of metabolic syndrome and urolithiasis. Int J Endocrinol 2015; 2015:570674. [PMID: 25873954 PMCID: PMC4385647 DOI: 10.1155/2015/570674] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/21/2015] [Accepted: 02/24/2015] [Indexed: 12/11/2022] Open
Abstract
There has been an increasing prevalence of kidney stones over the last 2 decades worldwide. Many studies have indicated a possible association between metabolic syndrome and kidney stone disease, particularly in overweight and obese patients. Many different definitions of metabolic syndrome have been suggested by various organizations, although the definition by the International Diabetes Federation (IDF) is universally considered as the most acceptable definition. The IDF definition revolves around 4 core components: obesity, dyslipidemia, hypertension, and diabetes mellitus. Several hypotheses have been proposed to explain the pathophysiology of urolithiasis resulting from metabolic syndrome, amongst which are the insulin resistance and Randall's plaque hypothesis. Similarly the pathophysiology of calcium and uric acid stone formation has been investigated to determine a connection between the two conditions. Studies have found many factors contributing to urolithiasis in patients suffering from metabolic syndrome, out of which obesity, overweight, and sedentary lifestyles have been identified as major etiological factors. Primary and secondary prevention methods therefore tend to revolve mainly around lifestyle improvements, including dietary and other preventive measures.
Collapse
Affiliation(s)
- Yee V. Wong
- Department of Urology, University Hospital Southampton NHS Trust, Southampton SO16 6YD, UK
| | - Paul Cook
- Department of Biochemical Pathology, University Hospital Southampton NHS Trust, Southampton SO16 6YD, UK
| | - Bhaskar K. Somani
- Department of Urology, University Hospital Southampton NHS Trust, Southampton SO16 6YD, UK
- *Bhaskar K. Somani:
| |
Collapse
|
21
|
Kernan WN, Ovbiagele B, Black HR, Bravata DM, Chimowitz MI, Ezekowitz MD, Fang MC, Fisher M, Furie KL, Heck DV, Johnston SCC, Kasner SE, Kittner SJ, Mitchell PH, Rich MW, Richardson D, Schwamm LH, Wilson JA. Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2014; 45:2160-236. [PMID: 24788967 DOI: 10.1161/str.0000000000000024] [Citation(s) in RCA: 2910] [Impact Index Per Article: 264.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this updated guideline is to provide comprehensive and timely evidence-based recommendations on the prevention of future stroke among survivors of ischemic stroke or transient ischemic attack. The guideline is addressed to all clinicians who manage secondary prevention for these patients. Evidence-based recommendations are provided for control of risk factors, intervention for vascular obstruction, antithrombotic therapy for cardioembolism, and antiplatelet therapy for noncardioembolic stroke. Recommendations are also provided for the prevention of recurrent stroke in a variety of specific circumstances, including aortic arch atherosclerosis, arterial dissection, patent foramen ovale, hyperhomocysteinemia, hypercoagulable states, antiphospholipid antibody syndrome, sickle cell disease, cerebral venous sinus thrombosis, and pregnancy. Special sections address use of antithrombotic and anticoagulation therapy after an intracranial hemorrhage and implementation of guidelines.
Collapse
|
22
|
Esposito K, Capuano A, Giugliano D. Metabolic syndrome and cancer: holistic or reductionist? Endocrine 2014; 45:362-4. [PMID: 24065310 DOI: 10.1007/s12020-013-0056-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 09/04/2013] [Indexed: 12/20/2022]
Abstract
Metabolic syndrome has become a major public health problem worldwide and represents a common clinical condition in countries with a high incidence of obesity and western dietary patterns. Metabolic syndrome associates with common cancers at many sites, including liver, colorectal, and bladder cancers in men, and endometrial, pancreatic, breast post-menopausal, and colorectal cancers in women. However, the role played by each single component of the syndrome on cancer risk is still unclear. For endometrial cancer, obesity and/or high circumference waist explain all the risk associated with the full metabolic syndrome, while for post-menopausal breast cancer, the risk conveyed by metabolic syndrome appears to be greater than its parts, as no single component explains the full risk associated with the syndrome. Future research should cover other avenues in order to elucidate the complexity of biological processes linking metabolic syndrome and cancer.
Collapse
Affiliation(s)
- Katherine Esposito
- Department of Clinical and Experimental Medicine, Second University of Naples, Naples, Italy
| | | | | |
Collapse
|
23
|
Xiao S, Fei N, Pang X, Shen J, Wang L, Zhang B, Zhang M, Zhang X, Zhang C, Li M, Sun L, Xue Z, Wang J, Feng J, Yan F, Zhao N, Liu J, Long W, Zhao L. A gut microbiota-targeted dietary intervention for amelioration of chronic inflammation underlying metabolic syndrome. FEMS Microbiol Ecol 2013; 87:357-67. [PMID: 24117923 PMCID: PMC4255291 DOI: 10.1111/1574-6941.12228] [Citation(s) in RCA: 273] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/14/2013] [Accepted: 09/17/2013] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation induced by endotoxin from a dysbiotic gut microbiota contributes to the development of obesity-related metabolic disorders. Modification of gut microbiota by a diet to balance its composition becomes a promising strategy to help manage obesity. A dietary scheme based on whole grains, traditional Chinese medicinal foods, and prebiotics (WTP diet) was designed to meet human nutritional needs as well as balance the gut microbiota. Ninety-three of 123 central obese volunteers (BMI ≥ 28 kg m(-2) ) completed a self-controlled clinical trial consisting of 9-week intervention on WTP diet followed by a 14-week maintenance period. The average weight loss reached 5.79 ± 4.64 kg (6.62 ± 4.94%), in addition to improvement in insulin sensitivity, lipid profiles, and blood pressure. Pyrosequencing of fecal samples showed that phylotypes related to endotoxin-producing opportunistic pathogens of Enterobacteriaceae and Desulfovibrionaceae were reduced significantly, while those related to gut barrier-protecting bacteria of Bifidobacteriaceae increased. Gut permeability, measured as lactulose/mannitol ratio, was decreased compared with the baseline. Plasma endotoxin load as lipopolysaccharide-binding protein was also significantly reduced, with concomitant decrease in tumor necrosis factor-α, interleukin-6, and an increase in adiponectin. These results suggest that modulation of the gut microbiota via dietary intervention may enhance the intestinal barrier integrity, reduce circulating antigen load, and ultimately ameliorate the inflammation and metabolic phenotypes.
Collapse
Affiliation(s)
- Shuiming Xiao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Effects of recommendations to follow the Dietary Approaches to Stop Hypertension (DASH) diet v. usual dietary advice on childhood metabolic syndrome: a randomised cross-over clinical trial. Br J Nutr 2013; 110:2250-9. [DOI: 10.1017/s0007114513001724] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The effects of the Dietary Approaches to Stop Hypertension (DASH) eating plan on childhood metabolic syndrome (MetS) and insulin resistance remain to be determined. The present study aimed to assess the effects of recommendations to follow the DASH diet v. usual dietary advice (UDA) on the MetS and its features in adolescents. In this randomised cross-over clinical trial, sixty post-pubescent adolescent girls with the MetS were randomly assigned to receive either the recommendations to follow the DASH diet or UDA for 6 weeks. After a 4-week washout period, the participants were crossed over to the alternate arm. The DASH group was recommended to consume a diet rich in fruits, vegetables and low-fat dairy products and low in saturated fats, total fats and cholesterol. UDA consisted of general oral advice and written information about healthy food choices based on healthy MyPlate. Compliance was assessed through the quantification of plasma vitamin C levels. In both the groups, fasting venous blood samples were obtained at baseline and at the end of each phase of the intervention. The mean age and weight of the participants were 14·2 (sd 1·7) years and 69 (sd 14·5) kg, respectively. Their mean BMI and waist circumference were 27·3 kg/m2 and 85·6 cm, respectively. Serum vitamin C levels tended to be higher in the DASH phase than in the UDA phase (860 (se 104) v. 663 (se 76) ng/l, respectively, P= 0·06). Changes in weight, waist circumference and BMI were not significantly different between the two intervention phases. Although changes in systolic blood pressure were not statistically significant between the two groups (P= 0·13), recommendations to follow the DASH diet prevented the increase in diastolic blood pressure compared with UDA (P= 0·01). We found a significant within-group decrease in serum insulin levels (101·4 (se 6·2) v. 90·0 (se 5·5) pmol/l, respectively, P= 0·04) and a non-significant reduction in the homeostasis model assessment for insulin resistance score (P= 0·12) in the DASH group. Compared with the UDA group, the DASH group experienced a significant reduction in the prevalence of the MetS and high blood pressure. Recommendations to follow the DASH eating pattern for 6 weeks among adolescent girls with the MetS led to reduced prevalence of high blood pressure and the MetS and improved diet quality compared with UDA. This type of healthy diet can be considered as a treatment modality for the MetS and its components in children.
Collapse
|
25
|
Lushchak OV, Gospodaryov DV, Rovenko BM, Yurkevych IS, Perkhulyn NV, Lushchak VI. Specific Dietary Carbohydrates Differentially Influence the Life Span and Fecundity of Drosophila melanogaster. ACTA ACUST UNITED AC 2013; 69:3-12. [DOI: 10.1093/gerona/glt077] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Neuhouser ML, Howard B, Lu J, Tinker LF, Van Horn L, Caan B, Rohan T, Stefanick ML, Thomson CA. A low-fat dietary pattern and risk of metabolic syndrome in postmenopausal women: the Women's Health Initiative. Metabolism 2012; 61:1572-81. [PMID: 22633601 PMCID: PMC3430820 DOI: 10.1016/j.metabol.2012.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 04/04/2012] [Accepted: 04/04/2012] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Nutrition plays an important role in metabolic syndrome etiology. We examined whether the Women's Health Initiative (WHI) Dietary Modification Trial influenced metabolic syndrome risk. MATERIALS/METHODS 48,835 postmenopausal women aged 50-79 years were randomized to a low-fat (20% energy from fat) diet (intervention) or usual diet (comparison) for a mean of 8.1 years. Blood pressure, waist circumference and fasting blood measures of glucose, HDL-cholesterol and triglycerides were measured on a subsample (n=2816) at baseline and years 1, 3 and 6 post-randomization. Logistic regression estimated associations of the intervention with metabolic syndrome risk and use of cholesterol-lowering and hypertension medications. Multivariate linear regression tested associations between the intervention and metabolic syndrome components. RESULTS At year 3, but not years 1 or 6, women in the intervention group (vs. comparison) had a non-statistically significant lower risk of metabolic syndrome (OR=0.83, 95%CI 0.59-1.18). Linear regression models simultaneously modeling the five metabolic syndrome components revealed significant associations of the intervention with metabolic syndrome at year 1 (p<0.0001), but not years 3 (p=0.19) and 6 (p=0.17). Analyses restricted to intervention-adherent participants strengthened associations at years 3 (p=0.05) and 6 (p=0.06). Cholesterol-lowering and hypertension medication use was 19% lower at year 1 for intervention vs. comparison group women (OR=0.81, 95% CI 0.60-1.09).Over the entire trial, fewer intervention vs. comparison participants used these medications (26.0% vs. 29.9%), although results were not statistically significant (p=0.89). CONCLUSIONS The WHI low-fat diet may influence metabolic syndrome risk and decrease use of hypertension and cholesterol-lowering medications. Findings have potential for meaningful clinical translation.
Collapse
Affiliation(s)
- Marian L Neuhouser
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Esposito K, Chiodini P, Colao A, Lenzi A, Giugliano D. Metabolic syndrome and risk of cancer: a systematic review and meta-analysis. Diabetes Care 2012; 35:2402-11. [PMID: 23093685 PMCID: PMC3476894 DOI: 10.2337/dc12-0336] [Citation(s) in RCA: 819] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Available evidence supports the emerging hypothesis that metabolic syndrome may be associated with the risk of some common cancers. We did a systematic review and meta-analysis to assess the association between metabolic syndrome and risk of cancer at different sites. RESEARCH DESIGN AND METHODS We conducted an electronic search for articles published through October 2011 without restrictions and by reviewing reference lists from retrieved articles. Every included study was to report risk estimates with 95% CIs for the association between metabolic syndrome and cancer. RESULTS We analyzed 116 datasets from 43 articles, including 38,940 cases of cancer. In cohort studies in men, the presence of metabolic syndrome was associated with liver (relative risk 1.43, P < 0.0001), colorectal (1.25, P < 0.001), and bladder cancer (1.10, P = 0.013). In cohort studies in women, the presence of metabolic syndrome was associated with endometrial (1.61, P = 0.001), pancreatic (1.58, P < 0.0001), breast postmenopausal (1.56, P = 0.017), rectal (1.52, P = 0.005), and colorectal (1.34, P = 0.006) cancers. Associations with metabolic syndrome were stronger in women than in men for pancreatic (P = 0.01) and rectal (P = 0.01) cancers. Associations were different between ethnic groups: we recorded stronger associations in Asia populations for liver cancer (P = 0.002), in European populations for colorectal cancer in women (P = 0.004), and in U.S. populations (whites) for prostate cancer (P = 0.001). CONCLUSIONS Metabolic syndrome is associated with increased risk of common cancers; for some cancers, the risk differs betweens sexes, populations, and definitions of metabolic syndrome.
Collapse
Affiliation(s)
- Katherine Esposito
- Department of Cardio-Thoracic and Respiratory Sciences, Second University of Naples, Naples, Italy
| | | | | | | | | |
Collapse
|
28
|
Lindholm CR, Ertel RL, Bauwens JD, Schmuck EG, Mulligan JD, Saupe KW. A high-fat diet decreases AMPK activity in multiple tissues in the absence of hyperglycemia or systemic inflammation in rats. J Physiol Biochem 2012; 69:165-75. [PMID: 22941749 DOI: 10.1007/s13105-012-0199-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 07/19/2012] [Indexed: 01/18/2023]
Abstract
Consumption of a high-fat diet (HFD) in experimental animal models initiates a series of molecular events and outcomes, including insulin resistance and obesity, that mimic the metabolic syndrome in humans. The relationship among, and order of, the molecular events linking a diet high in fat to pathologies is often unclear. In the present study, we provide several novel insights into the relationship between a HFD and AMP-activated protein kinase (AMPK), a key regulator of cellular metabolism and whole-body energy balance. HFD substantially decreased the activities of both isoforms of AMPK in white adipose tissue, heart, and liver. These decreases in AMPK activity occurred in the absence of decreased AMPK transcription, systemic inflammation, hyperglycemia, or elevated levels of free fatty acids. The HFD-induced decrease in AMPK activity was associated with systemic insulin resistance and hyperleptinemia. In blood, >98 % of AMPK activity was localized in agranulocytes as the α1 isoform. In contrast to the solid tissues studied, AMPK activities were not altered by HFD in granulocytes or agranulocytes. We conclude that HFD-induced obesity causes a broad, non-tissue, or isoform-specific lowering of AMPK activity. Given the central position AMPK plays in whole-body energy balance, this decreased AMPK activity may play a previously unrecognized role in obesity and its associated pathologies.
Collapse
Affiliation(s)
- Christopher R Lindholm
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin, 1630 Medical Sciences Center, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Metabolic syndrome (MetS) is a heterogeneous clinical entity represented by the occurrence of central obesity, hyperlipidaemia, hyperglycaemia and hypertension. The results of previous studies have shown that the probable common underlying pathophysiological factor for MetS is the insulin resistance phenomenon. However, the pathogenesis of the syndrome is still not well known. We present substantial information on MetS and the relationships between stroke and MetS as a compound entity, while individual components of MetS are well known risk factors for both first-in-life and recurrent ischaemic stroke. We also discuss primary and secondary stroke prevention in subjects with MetS.
Collapse
|
30
|
The effect of EPA and DHA on metabolic syndrome patients: a systematic review of randomised controlled trials. Br J Nutr 2012; 107 Suppl 2:S185-94. [DOI: 10.1017/s0007114512001572] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metabolic syndrome (MS) is characterised by accumulation of CVD risk factors. The use of very long chain n-3 polyunsaturated fatty acids (VLC n3 PUFA) could potentially benefit MS by reducing risk factors. To better understand the possible VLC n3 PUFA benefits, the literature was systematically reviewed for randomised controlled trials (RCT) that published effects of VLC n3 PUFA on MS patients. 17 RCT fulfilled the inclusion criteria and were analysed for relevance to the research question. The available RCT convincingly show that the administration of VLC n3 PUFA doses > 1 g for at least 3 months produces a significant reduction of triglycerides ranging from 7 % to 25 %. These results confirm the hypotriglyceridemic effect of VLC n3 PUFA in MS patients. The triglyceride lowering may produce further benefits by reducing the % of pro-atherogenic small dense LDL particles (sdLDL) and also perhaps by ameliorating the inflammatory process associated with MS. High doses of VLC n3 PUFA ( ≥ 3 g/day) may produce further TAG reductions but could raise other risk factors such as LDL-C. No clear effects were found on other MS markers. The combination of VLC n3 PUFA plus a statin may be useful to prevent the occurrence of coronary events. More studies are needed using different amounts of VLC n3 PUFA, time lengths, dietary backgrounds and different profiles of MS patients before clear recommendations can be made.
Collapse
|
31
|
Merriam PA, Persuitte G, Olendzki BC, Schneider K, Pagoto SL, Palken JL, Ockene IS, Ma Y. Dietary intervention targeting increased fiber consumption for metabolic syndrome. J Acad Nutr Diet 2012; 112:621-3. [PMID: 22709766 DOI: 10.1016/j.jand.2012.01.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 12/13/2011] [Indexed: 11/19/2022]
Affiliation(s)
- Philip A Merriam
- Division of Preventive and Behavioral Medicine, Department of Medicine, University of Massachusetts Medical School, 55 Lake Ave North, Worcester, MA 01655, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
The metabolic syndrome and risk of chronic kidney disease: pathophysiology and intervention strategies. J Nutr Metab 2012; 2012:652608. [PMID: 22523674 PMCID: PMC3317133 DOI: 10.1155/2012/652608] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 12/14/2011] [Accepted: 12/14/2011] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is characterized by a clustering of cardiovascular risk factors, including abdominal obesity, elevated blood pressure and glucose concentrations, and dyslipidemia. The presence of this clinical entity is becoming more pervasive throughout the globe as the prevalence of obesity increases worldwide. Moreover, there is increased recognition of the complications and mortality related to this syndrome. This paper looks to examine the link between metabolic syndrome and the development of chronic kidney disease.
Collapse
|
33
|
Goto T, Kim YI, Funakoshi K, Teraminami A, Uemura T, Hirai S, Lee JY, Makishima M, Nakata R, Inoue H, Senju H, Matsunaga M, Horio F, Takahashi N, Kawada T. Farnesol, an isoprenoid, improves metabolic abnormalities in mice via both PPARα-dependent and -independent pathways. Am J Physiol Endocrinol Metab 2011; 301:E1022-32. [PMID: 21862726 DOI: 10.1152/ajpendo.00061.2011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) control energy homeostasis. In this study, we showed that farnesol, a naturally occurring ligand of PPARs, could ameliorate metabolic diseases. Obese KK-Ay mice fed a high-fat diet (HFD) containing 0.5% farnesol showed significantly decreased serum glucose level, glucosuria incidence, and hepatic triglyceride contents. Farnesol-containing HFD upregulated the mRNA expressions of PPARα target genes involved in fatty acid oxidation in the liver. On the other hand, farnesol was not effective in upregulating the mRNA expressions of PPARγ target genes in white adipose tissues. Experiments using PPARα-deficient [(-/-)] mice revealed that the upregulation of fatty acid oxidation-related genes required PPARα function, but the suppression of hepatic triglyceride accumulation was partially PPARα-dependent. In hepatocytes isolated from the wild-type and PPARα (-/-) mice, farnesol suppressed triglyceride synthesis. In luciferase assay, farnesol activated both PPARα and the farnesoid X receptor (FXR) at similar concentrations. Moreover, farnesol increased the mRNA expression level of a small heterodimer partner known as one of the FXR target genes and decreased those of sterol regulatory element-binding protein-1c and fatty acid synthase in both the wild-type and PPARα (-/-) hepatocytes. These findings suggest that farnesol could improve metabolic abnormalities in mice via both PPARα-dependent and -independent pathways and that the activation of FXR by farnesol might contribute partially to the PPARα-independent hepatic triglyceride content-lowering effect. To our knowledge, this is the first study on the effect of the dual activators of PPARα and FXR on obesity-induced metabolic disorders.
Collapse
Affiliation(s)
- Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
There has been considerable interest in the possible association between impaired glucose tolerance (IGT) and neuropathy. A systematic literature search (Medline) and review was here performed. Twenty-three studies were evaluated. Fourteen investigated for the presence of neuropathy in patients with IGT. Nine studied patients with chronic idiopathic axonal polyneuropathy (CIAP), for the prevalence of IGT. The findings suggest that a significant proportion of patients with IGT may have neuropathy, particularly of the small-fibre and painful type. Similarly, a significant percentage of patients without another identifiable cause for painful axonal neuropathy may have IGT. This may however not be applicable to all populations. There are issues relating to the reproducibility, reliability and timing of a single glucose tolerance test (GTT) in establishing a diagnosis of IGT. Furthermore, it is possible neuropathic damage may occur at lower glucose levels than those defining IGT. In conclusion, further prospective long-term study of large IGT cohorts with known prestudy IGT duration is required to confirm and answer the many remaining questions about this presumed association. However, at present time, consideration of IGT as potential cause of painful small-fibre neuropathy appears justified, especially as patients may benefit from dietary and physical exercise interventions.
Collapse
Affiliation(s)
- Y A Rajabally
- Neuromuscular Clinic, Department of Neurology, University Hospitals of Leicester, UK.
| |
Collapse
|
35
|
Sikalidis AK, Varamini B. Roles of hormones and signaling molecules in describing the relationship between obesity and colon cancer. Pathol Oncol Res 2011; 17:785-90. [PMID: 21221874 DOI: 10.1007/s12253-010-9352-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 12/17/2010] [Indexed: 01/08/2023]
Abstract
Colon cancer represents a highly prevalent disease in the Western world. While dietary and lifestyle recommendations remain important factors in disease prevention and treatment, epidemiological data have made it clear that obesity and excess body weight remain significant risk factors for the disease. A number of potential direct and indirect relationships exist between obesity and increased risk of colon cancer. Several mechanisms which appear promising and warrant further investigation are discussed here, specifically the modifying role of insulin and insulin-like growth factors, leptin, adipose-tissue induced changes in estrogens and androgens, and inflammatory molecules. A brief review of these hormones and signaling molecules and their action in colon cancer development is described. A thorough integration and understanding of the mechanisms of action these systems exert on colonic epithelia will be important in designing studies and experiments aimed at elucidating disease etiology for prevention and treatment.
Collapse
Affiliation(s)
- Angelos K Sikalidis
- Division of Nutritional Sciences, Cornell University, 214 Savage Hall, Ithaca, NY 14853, USA.
| | | |
Collapse
|
36
|
Why and How Meet n-3 PUFA Dietary Recommendations? Gastroenterol Res Pract 2010; 2011:364040. [PMID: 21197079 PMCID: PMC3004387 DOI: 10.1155/2011/364040] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 09/20/2010] [Accepted: 10/09/2010] [Indexed: 01/12/2023] Open
Abstract
Obesity and the metabolic syndrome are systemic inflammatory diseases reaching epidemic proportions. Contemporary changes in human nutrition occurred characterized by increased consumption of fat and of vegetable oils rich in n-6 polyunsaturated fatty acids (PUFAs) together with decrease in n-3 PUFA-rich foods, resulting in an n-6/n-3 ratio of 10–20/1 in Western diet for a ratio around 1/1 in the diet of our ancestors. The literature provides compelling evidence for the health benefit of n-3 PUFA consumption on inflammation and metabolic syndrome prevention and treatment. Such evidence led to the establishment of comprehensive recommendations. However, we show here that, both in collective catering proposed to children and in hospital diet, it is not straightforward to meet such recommendations. Willingness of governments to institute changes, with accountable decisions on catering, nutritional education, and food processing, is required to face our neglected responsibility in promoting balanced diet and consumption of foods rich in essential nutrients in the general population.
Collapse
|
37
|
Graves BW. The obesity epidemic: scope of the problem and management strategies. J Midwifery Womens Health 2010; 55:568-78. [PMID: 20974419 DOI: 10.1016/j.jmwh.2010.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 04/01/2010] [Accepted: 05/10/2010] [Indexed: 01/08/2023]
Abstract
As the obesity epidemic increases, primary care clinicians are encountering obesity and health problems associated with obesity more frequently than ever before. In 2007, 41% of women were classified as obese, with a body mass index (BMI) of 30 or higher. Non-Hispanic blacks and Hispanics are more likely to be obese than white, non-Hispanics. A wide spectrum of health problems has been associated with obesity, including cardiovascular disease, diabetes, metabolic syndrome, osteoarthritis, and polycystic ovary syndrome. Obesity has been shown to be a low-grade inflammatory state, which may be responsible for many of the comorbidities. The general consensus recommends screening for obesity and counseling to promote weight loss. In some cases, pharmacotherapy and or bariatric surgery may be recommended.
Collapse
Affiliation(s)
- Barbara W Graves
- Midwifery Education Program, Baystate Medical Center, Springfield, MA 01199, USA.
| |
Collapse
|
38
|
Diet and plasma evaluation of the main isomers of conjugated linoleic acid and trans-fatty acids in a population sample from Mediterranean north-east Spain. Food Chem 2010. [DOI: 10.1016/j.foodchem.2010.04.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
39
|
Furie KL, Kasner SE, Adams RJ, Albers GW, Bush RL, Fagan SC, Halperin JL, Johnston SC, Katzan I, Kernan WN, Mitchell PH, Ovbiagele B, Palesch YY, Sacco RL, Schwamm LH, Wassertheil-Smoller S, Turan TN, Wentworth D. Guidelines for the prevention of stroke in patients with stroke or transient ischemic attack: a guideline for healthcare professionals from the american heart association/american stroke association. Stroke 2010; 42:227-76. [PMID: 20966421 DOI: 10.1161/str.0b013e3181f7d043] [Citation(s) in RCA: 1135] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this updated statement is to provide comprehensive and timely evidence-based recommendations on the prevention of ischemic stroke among survivors of ischemic stroke or transient ischemic attack. Evidence-based recommendations are included for the control of risk factors, interventional approaches for atherosclerotic disease, antithrombotic treatments for cardioembolism, and the use of antiplatelet agents for noncardioembolic stroke. Further recommendations are provided for the prevention of recurrent stroke in a variety of other specific circumstances, including arterial dissections; patent foramen ovale; hyperhomocysteinemia; hypercoagulable states; sickle cell disease; cerebral venous sinus thrombosis; stroke among women, particularly with regard to pregnancy and the use of postmenopausal hormones; the use of anticoagulation after cerebral hemorrhage; and special approaches to the implementation of guidelines and their use in high-risk populations.
Collapse
|
40
|
Esposito K, Giugliano D. Mediterranean diet and the metabolic syndrome: the end of the beginning. Metab Syndr Relat Disord 2010; 8:197-200. [PMID: 20158447 DOI: 10.1089/met.2009.0095] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The metabolic syndrome is now both a public health and a clinical problem. The most recent estimates from the National Health and Nutrition Examination Survey (NHANES) 2003-2006 reported that approximately 34% of adults in the United States have the metabolic syndrome. The relationship between adherence to a Mediterranean diet and the prevalence of metabolic syndrome has been explored in cross-sectional studies with discordant results. Two prospective studies with a follow up of 6 and 7.5 years show that subjects with the highest adherence to a Mediterranean diet had lower cumulative incidence of metabolic syndrome than those with the lowest adherence. Two interventional trials document a positive effect of a Mediterranean diet on reversal of metabolic syndrome. The Mediterranean diet may be also useful to ameliorate liver function tests in obese patients with insulin resistance and diabetes. There is no consensus as to what diet is the optimal one for patients who have the metabolic syndrome. A Mediterranean diet that is moderately lower in carbohydrate (45%), and moderately higher in fat (35-40%), with less than 10% of saturated fat, may be beneficial for ameliorating features of the metabolic syndrome, including effects on insulin sensitivity, blood lipids, and liver function.
Collapse
Affiliation(s)
- Katherine Esposito
- Department of Geriatrics and Metabolic Diseases, Second University of Naples, Italy.
| | | |
Collapse
|
41
|
Jiménez-Gómez Y, Marín C, Peérez-Martínez P, Hartwich J, Malczewska-Malec M, Golabek I, Kiec-Wilk B, Cruz-Teno C, Rodríguez F, Gómez P, Gómez-Luna MJ, Defoort C, Gibney MJ, Pérez-Jiménez F, Roche HM, López-Miranda J. A low-fat, high-complex carbohydrate diet supplemented with long-chain (n-3) fatty acids alters the postprandial lipoprotein profile in patients with metabolic syndrome. J Nutr 2010; 140:1595-601. [PMID: 20631323 DOI: 10.3945/jn.109.120816] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Dietary fat intake plays a critical role in the development of metabolic syndrome (MetS). This study addressed the hypothesis that dietary fat quantity and quality may differentially modulate postprandial lipoprotein metabolism in MetS patients. A multi-center, parallel, randomized, controlled trial conducted within the LIPGENE study randomly assigned MetS patients to 1 of 4 diets: high-SFA [HSFA; 38% energy (E) from fat, 16% E as SFA], high-monounsaturated fatty acid [HMUFA; 38% E from fat, 20% E as MUFA], and 2 low-fat, high-complex carbohydrate [LFHCC; 28% E from fat] diets supplemented with 1.24 g/d of long-chain (LC) (n-3) PUFA (ratio 1.4 eicosapentaenoic acid:1 docosahexaenoic acid) or placebo (1.24 g/d of high-oleic sunflower-seed oil) for 12 wk each. A fat challenge with the same fat composition as the diets was conducted pre- and postintervention. Postprandial total cholesterol, triglycerides (TG), apolipoprotein (apo) B, apo B-48, apo A-I, LDL-cholesterol, HDL-cholesterol and cholesterol, TG, retinyl palmitate, and apo B in TG-rich lipoproteins (TRL; large and small) were determined pre- and postintervention. Postintervention, postprandial TG (P < 0.001) and large TRL-TG (P = 0.009) clearance began earlier and was faster in the HMUFA group compared with the HSFA and LFHCC groups. The LFHCC (n-3) group had a lower postprandial TG concentration (P < 0.001) than the other diet groups. Consuming the LFHCC diet increased the TG (P = 0.04), large TRL-TG (P = 0.01), TRL-cholesterol (P < 0.001), TRL-retinyl palmitate (P = 0.001), and TRL-apo B (P = 0.002) area under the curve compared with preintervention values. In contrast, long-term ingestion of the LFHCC (n-3) diet did not augment postprandial TG and TRL metabolism. In conclusion, postprandial abnormalities associated with MetS can be attenuated with LFHCC (n-3) and HMUFA diets. The adverse postprandial TG-raising effects of long-term LFHCC diets may be avoided by concomitant LC (n-3) PUFA supplementation to weight-stable MetS patients.
Collapse
Affiliation(s)
- Yolanda Jiménez-Gómez
- Reina Sofía University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abete I, Astrup A, Martínez JA, Thorsdottir I, Zulet MA. Obesity and the metabolic syndrome: role of different dietary macronutrient distribution patterns and specific nutritional components on weight loss and maintenance. Nutr Rev 2010; 68:214-31. [PMID: 20416018 DOI: 10.1111/j.1753-4887.2010.00280.x] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Weight loss and subsequent body weight maintenance are difficult for obese individuals despite the wide variety of dietary regimens and approaches. A substantial body of scientific evidence has shown that by simply varying the macronutrient distribution and composition of dietary factors, weight losses of varying amounts, longer-term body weight maintenance periods, better appetite regulation, and changes in features of the metabolic syndrome can be achieved. At present, renewed efforts are underway to increase the protein content of weight-loss diets, simultaneously restrict fat consumption to no more than 30%, favor polyunsaturated fat, have carbohydrates account for between 40 and 50% of total energy intake, and promote the consumption of low-glycemic foods. The present article reviews the scientific evidence for the effects of several dietary manipulations and sustainable strategies for weight loss and body weight stability as well as for treating specific features of the metabolic syndrome.
Collapse
Affiliation(s)
- Itziar Abete
- Department of Nutrition, Food Science, Physiology and Toxicology, University of Navarra, Pamplona, Navarra, Spain
| | | | | | | | | |
Collapse
|
43
|
Various Terpenoids Derived from Herbal and Dietary Plants Function as PPAR Modulators and Regulate Carbohydrate and Lipid Metabolism. PPAR Res 2010; 2010:483958. [PMID: 20613991 PMCID: PMC2896613 DOI: 10.1155/2010/483958] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 03/23/2010] [Indexed: 12/14/2022] Open
Abstract
Several herbal plants improve medical conditions. Such plants contain many bioactive phytochemicals. Terpenoids (also called “isoprenoids”) constitute one of the largest families of natural products accounting for more than 40,000 individual compounds of both primary and secondary metabolisms. In particular, terpenoids are contained in many herbal plants, and several terpenoids have been shown to be available for pharmaceutical applications, for example, artemisinin and taxol as malaria and cancer medicines, respectively. Various terpenoids are contained in many plants for not only herbal use but also dietary use. In this paper, we describe several bioactive terpenoids contained in herbal or dietary plants, which can modulate the activities of ligand-dependent transcription factors, namely, peroxisome proliferator-activated receptors (PPARs). Because PPARs are dietary lipid sensors that control energy homeostasis, daily eating of these terpenoids might be useful for the management for obesity-induced metabolic disorders, such as type 2 diabetes, hyperlipidemia, insulin resistance, and cardiovascular diseases.
Collapse
|
44
|
van Dijk SJ, Feskens EJM, Bos MB, Hoelen DWM, Heijligenberg R, Bromhaar MG, de Groot LCPGM, de Vries JHM, Müller M, Afman LA. A saturated fatty acid-rich diet induces an obesity-linked proinflammatory gene expression profile in adipose tissue of subjects at risk of metabolic syndrome. Am J Clin Nutr 2009; 90:1656-64. [PMID: 19828712 DOI: 10.3945/ajcn.2009.27792] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Changes in dietary fat composition could lower the risk of developing metabolic syndrome. Adipose tissue is an interesting tissue in this respect because of its role in lipid metabolism and inflammation. OBJECTIVE Our objective was to investigate the effect of a saturated fatty acid (SFA)- and a monounsaturated fatty acid (MUFA)-rich diet on insulin sensitivity, serum lipids, and gene expression profiles of adipose tissue in subjects at risk of metabolic syndrome. DESIGN A parallel controlled-feeding trial was conducted in 20 abdominally overweight subjects. Subjects received an SFA diet or a MUFA diet for 8 wk. Plasma and subcutaneous adipose tissue samples were obtained, and insulin sensitivity was measured by using a hyperinsulinemic-euglycemic clamp. Adipose tissue samples underwent whole-genome microarray and histologic analysis. Plasma and adipose tissue fatty acid composition and concentrations of serum cholesterol and plasma cytokine were determined. RESULTS Consumption of the SFA diet resulted in increased expression of genes involved in inflammation processes in adipose tissue, without changes in morphology or insulin sensitivity. The MUFA diet led to a more antiinflammatory gene expression profile, which was accompanied by a decrease in serum LDL-cholesterol concentrations and an increase in plasma and adipose tissue oleic acid content. CONCLUSIONS Consumption of an SFA diet resulted in a proinflammatory "obesity-linked" gene expression profile, whereas consumption of a MUFA diet caused a more antiinflammatory profile. This suggests that replacement of dietary SFA with MUFA could prevent adipose tissue inflammation and may reduce the risk of inflammation-related diseases such as metabolic syndrome. This trial was registered at clinicaltrials.gov as NCT00405197.
Collapse
Affiliation(s)
- Susan J van Dijk
- The Division of Human Nutrition, Wageningen University, Wageningen, Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Esposito K, Maiorino MI, Di Palo C, Giugliano D. Adherence to a Mediterranean diet and glycaemic control in Type 2 diabetes mellitus. Diabet Med 2009; 26:900-7. [PMID: 19719711 DOI: 10.1111/j.1464-5491.2009.02798.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIMS Mediterranean-type diets reduce the risk of Type 2 diabetes. Whether a Mediterranean-type diet improves glycaemic control in diabetes remains unknown. METHODS We conducted a cross-sectional analysis in 901 outpatients with Type 2 diabetes attending diabetes clinics located in Campania County, South Italy. We explored the relation between glycated haemoglobin (HbA(1c)), measured centrally, self-measured pre- and postprandial glucose levels and consumption of a Mediterranean-type diet. Adherence to a Mediterranean-type diet was assessed by a 9-point scale that incorporated the salient characteristics of this diet (range of scores, 0-9, with higher scores indicating greater adherence). The study was conducted from 2001 to 2007. RESULTS Diabetic patients with the highest scores (6-9) had lower body mass index and waist circumferences, a lower prevalence of the metabolic syndrome and lower HbA(1c) and post-meal glucose levels than diabetic patients with the lowest scores (0-3). In multivariate analysis, mean HbA(1c) and 2-h post-meal glucose concentrations were significantly lower in diabetic patients with high adherence to a Mediterranean-type diet than those with low adherence [difference: HbA(1c) 0.9%, 95% confidence intervals (CI) 0.5-1.2%, P < 0.001; 2-h glucose 2.2 mmol/l, 95% CI 0.8-2.9 mmol/l, P < 0.001]. CONCLUSIONS In Type 2 diabetes, greater adherence to a Mediterranean-type diet is associated with lower HbA(1c) and postprandial glucose levels.
Collapse
Affiliation(s)
- K Esposito
- Department of Geriatrics and Metabolic Diseases, Second University of Naples, 80138 Naples, Italy
| | | | | | | |
Collapse
|
46
|
Hu GX, Chen GR, Xu H, Ge RS, Lin J. Activation of the AMP activated protein kinase by short-chain fatty acids is the main mechanism underlying the beneficial effect of a high fiber diet on the metabolic syndrome. Med Hypotheses 2009; 74:123-6. [PMID: 19665312 DOI: 10.1016/j.mehy.2009.07.022] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 07/08/2009] [Indexed: 12/20/2022]
Abstract
The metabolic syndrome, a cluster of conditions including abdominal obesity, insulin resistance, dyslipidemia, and elevated blood pressure, is a natural consequence of over nutrition and a sedentary lifestyle. The prevalence of the metabolic syndrome has increased to epidemic proportions in the world. The exact pathogenesis of the metabolic syndrome remains unclear, but it is known to be a complex interaction between genetic, metabolic, and environmental factors. Promotion of physical activity and dietary management are still the main methods for the prevention and management of the metabolic syndrome. Numerous experimental and clinical studies have demonstrated the beneficial effects of high fiber diet on the metabolic syndrome. The principal beneficial effects of a fiber-rich diet in these patients are: prevention of obesity, improved glucose levels, and control of the profile of blood lipids. Dietary fiber may also favor the control of arterial blood pressure. How dietary fiber exerts its beneficial effects on the metabolic syndrome is not well understood. AMP activated protein kinase (AMPK) functions as a major cellular fuel gauge and a master regulator of metabolic homeostasis. Several lines of evidence suggest that AMPK can be activated by short-chain fatty acids (SCFA) either directly or indirectly. It is our hypothesis that the main mechanism underlying the beneficial effect of a high fiber diet on the metabolic syndrome is the increased SCFA production in the colon leading to a higher concentration of SCFA in the portal vein, which activates the AMPK in the liver.
Collapse
Affiliation(s)
- Guo-Xin Hu
- Institute of Molecular Toxicology and Pharmacology, School of Pharmacy, Wenzhou Medical College, Wenzhou, Zhejiang 325000, PR China
| | | | | | | | | |
Collapse
|
47
|
Diet quality of a population sample from coastal north-east Spain evaluated by a Mediterranean adaptation of the diet quality index (DQI). Public Health Nutr 2009; 13:12-24. [PMID: 19545468 DOI: 10.1017/s1368980009990231] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To assess the adherence to the Mediterranean dietary pattern in the population from a coastal region from north-east Spain and its relationship to diseases, applying the Mediterranean Diet Quality Index (M-DQI) validated by the use of several biomarkers. DESIGN Cross-sectional nutrition survey. SETTING Population-based random sample derived from the Catalan Nutrition Survey. SUBJECTS A total of 621 healthy adults. RESULTS The Catalan representative sample presented a mean M-DQI score of 6.6 (sd 2.3, median 7, range 0-14). The percentage of adherence to the Mediterranean diet was 53 %; 10 % of subjects showed high adherence to the Mediterranean diet, while only 2 % were categorized as poorest adherence. The plasma fatty acid profile of the Catalan sample progressed with perfect regularity throughout the index ranges. Both EPA and DHA presented a significant correlation to the M-DQI (r = -0.410 for EPA and -0.360 for DHA). A significant increase in palmitic, oleic and alpha-linolenic acids and a significant decrease in stearic, linoleic and arachidonic acids content were also observed. The mean values for the M-DQI according to the clinical characteristics of the Catalan sample were also calculated. CONCLUSIONS The M-DQI has been demonstrated a suitable tool for assessment of an individual's nutritional status according to the Mediterranean dietary pattern and for clinical purposes. Although the current diet followed in Catalonia seems to agree with the main characteristics of the Mediterranean diet, the promotion of the Mediterranean pattern should be reinforced in the Catalan population, especially among young people.
Collapse
|
48
|
Marasini B, Massarotti M. What rheumatologists should know about gout and cardiovascular disease. J Rheumatol 2009; 36:854-5. [PMID: 19342726 DOI: 10.3899/jrheum.081039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Esposito K, Maiorino M, Giugliano D. Which diet is best for diabetes? Diabetologia 2009; 52:988-9. [PMID: 19221708 DOI: 10.1007/s00125-009-1292-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 01/22/2009] [Accepted: 01/22/2009] [Indexed: 10/21/2022]
|
50
|
Schwellnus MP, Patel DN, Nossel CJ, Dreyer M, Whitesman S, Derman EW. Healthy lifestyle interventions in general practice Part 6: Lifestyle and metabolic syndrome. S Afr Fam Pract (2004) 2009. [DOI: 10.1080/20786204.2009.10873841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|