1
|
Andres-Hernando A, Orlicky DJ, Kuwabara M, Fini MA, Tolan DR, Johnson RJ, Lanaspa MA. Activation of AMPD2 drives metabolic dysregulation and liver disease in mice with hereditary fructose intolerance. Commun Biol 2024; 7:849. [PMID: 38992061 PMCID: PMC11239681 DOI: 10.1038/s42003-024-06539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Hereditary fructose intolerance (HFI) is a painful and potentially lethal genetic disease caused by a mutation in aldolase B resulting in accumulation of fructose-1-phosphate (F1P). No cure exists for HFI and treatment is limited to avoid exposure to fructose and sugar. Using aldolase B deficient mice, here we identify a yet unrecognized metabolic event activated in HFI and associated with the progression of the disease. Besides the accumulation of F1P, here we show that the activation of the purine degradation pathway is a common feature in aldolase B deficient mice exposed to fructose. The purine degradation pathway is a metabolic route initiated by adenosine monophosphate deaminase 2 (AMPD2) that regulates overall energy balance. We demonstrate that very low amounts of fructose are sufficient to activate AMPD2 in these mice via a phosphate trap. While blocking AMPD2 do not impact F1P accumulation and the risk of hypoglycemia, its deletion in hepatocytes markedly improves the metabolic dysregulation induced by fructose and corrects fat and glycogen storage while significantly increasing the voluntary tolerance of these mice to fructose. In summary, we provide evidence for a critical pathway activated in HFI that could be targeted to improve the metabolic consequences associated with fructose consumption.
Collapse
Affiliation(s)
- Ana Andres-Hernando
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO, USA
| | - David J Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Masanari Kuwabara
- Department of Cardiology, Toranomon Hospital, Tokyo, Japan
- Division of Public Health, Center for Community Medicine, Jichi Medical University, Tochigi, Japan
| | - Mehdi A Fini
- Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Miguel A Lanaspa
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO, USA.
| |
Collapse
|
2
|
Mehdad S, Belghiti H, Zahrou FE, Guerinech H, Mouzouni FZ, El Hajjab A, El Berri H, El Ammari L, Benaich S, Benkirane H, Barkat A, Aguenaou H. Vitamin D status and its relationship with obesity indicators in Moroccan adult women. Nutr Health 2023; 29:673-681. [PMID: 35435056 DOI: 10.1177/02601060221094376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Background: Although vitamin D deficiency has been studied in various populations, there are few data on its prevalence and associated factors among Moroccan women. Aim: To determine the prevalence of vitamin D deficiency and investigate its association with body mass index (BMI), waist circumference, and serum concentrations of parathyroid hormone, calcium, and phosphorus in a sample of Moroccan adult women. Methods: This is a cross-sectional study conducted at Mohammed V Military Hospital of Instruction, Rabat. Anthropometric measurements and biochemical analyses were performed using standard procedures Results: A total of 714 women aged 18-65 years participated in this study. The overall prevalence of vitamin D deficiency was 74.4%. Approximately 24% and 51% of women had severe and moderate vitamin D deficiency, respectively. Serum 25-hydroxyvitamin D (25(OH)D) concentrations were inversely correlated with BMI in vitamin D-deficient subjects (P = 0.036) and with parathyroid hormone 1-84 (PTH1-84) levels in the study sample (P = 0.010). PTH1-84 concentrations were greater among overweight/obese individuals compared to their non-overweight peers (P = 0.001) and tended to be higher among vitamin D-deficient women than vitamin D-sufficient women (P = 0.053). Conclusion: This study showed a very high prevalence of vitamin D deficiency in this sample of Moroccan women. Lower serum 25(OH)D levels were associated with increased BMI in vitamin D-deficient women and with elevated PTH1-84 levels among the study sample. Although these findings come from a convenience sample of women that attended a nutrition clinic, they underscore the urgent need to develop public health interventions to improve women's vitamin D status.
Collapse
Affiliation(s)
- Slimane Mehdad
- Physiology and Physiopathology Research Team, Research Centre of Human Pathologies Genomics, Faculty of Sciences, Mohammed V University in Rabat, Morocco
- Clinical Nutrition Unit, Mohammed V Military Hospital of Instruction, Rabat, Morocco
- Ibn Tofail University- CNESTEN, Joint Unit of Nutrition, Health and Environment, Laboratory of Biology and Health, FSK, Regional Designated Center for Nutrition (AFRA/IAEA), Kenitra, 14000, Morocco
| | - Hakim Belghiti
- Clinical Nutrition Unit, Mohammed V Military Hospital of Instruction, Rabat, Morocco
- Ibn Tofail University- CNESTEN, Joint Unit of Nutrition, Health and Environment, Laboratory of Biology and Health, FSK, Regional Designated Center for Nutrition (AFRA/IAEA), Kenitra, 14000, Morocco
| | - Fatima Ezzahra Zahrou
- Ibn Tofail University- CNESTEN, Joint Unit of Nutrition, Health and Environment, Laboratory of Biology and Health, FSK, Regional Designated Center for Nutrition (AFRA/IAEA), Kenitra, 14000, Morocco
| | - Hassania Guerinech
- Clinical Nutrition Unit, Mohammed V Military Hospital of Instruction, Rabat, Morocco
| | | | - Amina El Hajjab
- Ibn Tofail University- CNESTEN, Joint Unit of Nutrition, Health and Environment, Laboratory of Biology and Health, FSK, Regional Designated Center for Nutrition (AFRA/IAEA), Kenitra, 14000, Morocco
| | | | | | - Souad Benaich
- Ibn Tofail University- CNESTEN, Joint Unit of Nutrition, Health and Environment, Laboratory of Biology and Health, FSK, Regional Designated Center for Nutrition (AFRA/IAEA), Kenitra, 14000, Morocco
| | - Hasnae Benkirane
- Ibn Tofail University- CNESTEN, Joint Unit of Nutrition, Health and Environment, Laboratory of Biology and Health, FSK, Regional Designated Center for Nutrition (AFRA/IAEA), Kenitra, 14000, Morocco
| | - Amina Barkat
- Health and Nutrition Research Team of the Mother-Child Couple, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Hassan Aguenaou
- Ibn Tofail University- CNESTEN, Joint Unit of Nutrition, Health and Environment, Laboratory of Biology and Health, FSK, Regional Designated Center for Nutrition (AFRA/IAEA), Kenitra, 14000, Morocco
| |
Collapse
|
3
|
Wu Q, Ye Z, Zhang Y, Yang S, Zhou C, Liu M, Zhang Y, Zhang Z, He P, Li R, Li H, Jiang J, Ai J, Nie J, Liu C, Qin X. A U-shaped association between dietary phosphorus intake and new-onset diabetes: A nationwide cohort study in China. Nutr Metab Cardiovasc Dis 2023; 33:1932-1940. [PMID: 37482482 DOI: 10.1016/j.numecd.2023.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND AND AIMS The association between dietary phosphorus intake and the risk of diabetes remains uncertain. We aimed to investigate the relation of dietary phosphorus intake with new-onset diabetes among Chinese adults. METHODS AND RESULTS A total of 16,272 participants who were free of diabetes at baseline from the China Health and Nutrition Survey were included. Dietary intake was measured by 3 consecutive 24-h dietary recalls combined with a household food inventory. Participants with self-reported physician-diagnosed diabetes, or fasting glucose ≥7.0 mmol/L or glycated hemoglobin ≥6.5% during the follow-up were defined as having new-onset diabetes. During a median follow-up of 9.0years, 1101 participants developed new-onset diabetes. Overall, the association between dietary phosphorus intake with new-onset diabetes followed a U-shape (P for nonlinearity<0.001). The risk of new-onset diabetes significantly decreased with the increment of dietary phosphorus intake (per SD increment: HR, 0.64; 95%CI, 0.48-0.84) in participants with phosphorus intake <921.6 mg/day, and increased with the increment of dietary phosphorus intake (per SD increment: HR, 1.33; 95%CI, 1.16-1.53) in participants with phosphorus intake ≥921.6 mg/day. Consistently, when dietary phosphorus intake was assessed as quintiles, compared with those in the 3rd quintile (905.0-<975.4 mg/day), significantly higher risks of new-onset diabetes were found in participants in the 1st-2nd quintiles (<905.0 mg/day: HR, 1.59; 95%CI, 1.30-1.94), and 4th-5th quintiles (≥975.4 mg/day: HR, 1.46; 95%CI, 1.19-1.78). CONCLUSIONS There was a U-shaped association between dietary phosphorus intake and new-onset diabetes in general Chinese adults, with an inflection point at 921.6 mg/day and a minimal risk at 905.0-975.4 mg/day of dietary phosphorus intake.
Collapse
Affiliation(s)
- Qimeng Wu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Ziliang Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Yanjun Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Sisi Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Chun Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Mengyi Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Zhuxian Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Panpan He
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Rui Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei 230032, China; Institute of Biomedicine, Anhui Medical University, Hefei 230032, China
| | - Huan Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Jianping Jiang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Jun Ai
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Jing Nie
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Chengzhang Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei 230032, China; Institute of Biomedicine, Anhui Medical University, Hefei 230032, China
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei 230032, China; Institute of Biomedicine, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
4
|
Ağbaht K, Pişkinpaşa SV. Serum TSH, 25(OH) D and phosphorus levels predict weight loss in individuals with diabetes/prediabetes and morbid obesity: a single-center retrospective cohort analysis. BMC Endocr Disord 2022; 22:282. [PMID: 36401211 PMCID: PMC9673446 DOI: 10.1186/s12902-022-01202-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 11/05/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND To evaluate the association of vitamin D and thyroid-stimulating hormone (TSH) with weight loss (WL) percentage (%) in patients with diabetes/prediabetes and Class II/III obesity. METHODS A retrospective cohort study was designed. Data were collected from a database of a referral endocrinology clinic that is prospectively and systematically generated. After exclusion of unavailable cases, the study enrolled 285 patients (51 ± 11 years old, female/male = 208/77; diabetes/prediabetes = 159/126; no/on levothyroxine replacement = 176/109; Class II/III obesity = 184/101, respectively) who maintained euthyroidism and were followed up for ≥6 months. The data were analyzed to determine the predictors of WL%. RESULTS Compared with baseline, in the median 22 months of follow-up, the whole study group lost 5.1% of their baseline body weight. As most obesity management trials define success as 'at least 10% of WL compared to baseline', we stratified the patients based on WL% extents. The distribution was as follow: Group 1 (n = 61) lost ≥10% body weight, Group 2 (n = 162) lost < 10% body weight, while Group 3 (n = 62) gained weight by the final visit. In groups 1 and 2 (weight losers), the serum thyroid stimulatig hormone (TSH) and parathyroid hormone (PTH) levels decreased and the free thyroxine (fT4), calcium, phosphorus, and 25-hydroxyvitamin D (25(OH)D) levels increased. In Group 3 (weight gainers), these changes were not observed (except for an increase in calcium levels). Regression analysis revealed that the final visit TSH (β = - 0.14, p < 0.05), 25(OH) D (β = 0.15, p < 0.05), and phosphorus (β = 0.20, p < 0.05) levels predicted WL%. However, if patients with autoimmune thyroiditis were excluded from the analysis, the decrease in TSH levels was not statistically significant. CONCLUSIONS Serum TSH, phosphorus, and 25(OH) D levels predict WL% in euthyroid patients with diabetes/prediabetes and morbid obesity. TSH predictivity seems to be a function of thyroid autoimmunity present with increased frequency in this cohort. Greater levels of phosphorus within the reference range and a sufficient vitamin D status are associated with a greater WL%.
Collapse
Affiliation(s)
- Kemal Ağbaht
- Defne Hospital, Endocrinology and Metabolic Diseases Department, Odabaşı Mahallesi, Uğur Mumcu Bulvarı, No: 101, Antakya, Hatay, Turkey.
| | | |
Collapse
|
5
|
Hoyt M, Song Y, Gao S, O'Palka J, Zhang J. Intake of Calcium, Magnesium, and Phosphorus and Risk of Pancreatic Cancer in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2022; 41:747-757. [PMID: 34586963 DOI: 10.1080/07315724.2021.1970047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/09/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022]
Abstract
ObjectiveFew epidemiological studies have investigated the associations between calcium, magnesium, and phosphorus intake and pancreatic cancer. We examined these associations in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial.MethodsDiet was assessed using the Dietary Questionnaire (DQX) at baseline in the intervention arm and the Dietary History Questionnaire (DHQ) in 1999 or around the third anniversary of randomization in both the intervention and control arms. During a median follow-up of 12.2 years, 279 cases of pancreatic cancer occurred from 58,477 participants who completed DQX; 380 cases arose from 101,622 participants who responded to DHQ over a median follow-up of 8.9 years. Cox proportional hazards regression was used to estimate hazard ratios (HR) and 95% confidence intervals (CI).ResultsTotal calcium intake was inversely associated with pancreatic cancer [HR (95% CI) for the fourth vs. the first quartiles in the DHQ cohort: 0.67 (0.47, 0.96); p-trend: 0.035]. An inverse association was also observed for total magnesium intake [HR (95% CI) for the fourth vs. the first quartiles in the DQX cohort: 0.61 (0.37, 1.00); p-trend: 0.023]. Reduced risk associated with total calcium intake was confined to subjects with a high fat intake (>73 g/day) in the DHQ cohort (p-interaction: 0.16).ConclusionsThere was not a significant association between dietary phosphorus intake and pancreatic cancer risk in both cohorts. Total intake of calcium and magnesium are associated with a lower pancreatic cancer risk. The effect of total calcium intake was modified by fat intake.
Collapse
Affiliation(s)
- Margaret Hoyt
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Yiqing Song
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Sujuan Gao
- Department of Biostatistics, Indiana University Richard M. Fairbanks School of Public Health and School of Medicine, Indianapolis, IN, USA
| | - Jacquelynn O'Palka
- Department of Nutrition and Dietetics, Indiana University School of Health and Human Sciences, Indianapolis, IN, USA
| | - Jianjun Zhang
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
6
|
Wong SK. A Review of Current Evidence on the Relationship between Phosphate Metabolism and Metabolic Syndrome. Nutrients 2022; 14:4525. [PMID: 36364791 PMCID: PMC9656201 DOI: 10.3390/nu14214525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 07/30/2023] Open
Abstract
Phosphorus, present as phosphate in biological systems, is an essential mineral for various biological activities and biochemical processes. Numerous studies have indicated that disturbed phosphate balance may contribute to the development of metabolic syndrome (MetS). However, no consistent result was found on the association between phosphorus intake and serum phosphate concentration with MetS. It is believed that both positive and negative impacts of phosphorus/phosphate co-exist in parallel during MetS condition. Reduced phosphate level contributed to the development of obesity and hyperglycaemia. Low phosphate is believed to compromise energy production, reduce exercise capacity, increase food ingestion, and impair glucose metabolism. On the other hand, the effects of phosphorus/phosphate on hypertension are rather complex depending on the source of phosphorus and subjects' health conditions. Phosphorus excess activates sympathetic nervous system, renin-angiotensin-aldosterone system, and induces hormonal changes under pathological conditions, contributing to the blood pressure-rising effects. For lipid metabolism, adequate phosphate content ensures a balanced lipid profile through regulation of fatty acid biosynthesis, oxidation, and bile acid excretion. In conclusion, phosphate metabolism serves as a potential key feature for the development and progression of MetS. Dietary phosphorus and serum phosphate level should be under close monitoring for the management of MetS.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
7
|
Zulkifli MF, Radzi MNFM, Saludes JP, Dalisay DS, Ismail WIW. Potential of Natural Honey in Controlling Obesity and its Related Complications. J Evid Based Integr Med 2022; 27:2515690X221103304. [PMID: 36263596 PMCID: PMC9585569 DOI: 10.1177/2515690x221103304] [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] [Indexed: 11/05/2022] Open
Abstract
Honey has a long history of therapeutic properties for multiple diseases, including inflammation and oxidative stress. This review aimed to provide a better understanding and renewed interest in the potential role of honey in obesity control, obesity-related diseases treatment and weight management, with specific reference to its components and the effect of honey overall. There is compelling evidence that honey possesses the desired properties for this purpose, as seen in the in vitro, in silico, in vivo and clinical analyses discussed in this review. This review also highlights the components potentially responsible for the health benefits of honey. Honey and its components reduce blood sugar levels, improve insulin sensitivity and lipid metabolism by reducing triglycerides, and reduce total cholesterol and LDL levels while increasing HDL levels that prevent excessive weight gain and reduce the risk of obesity and its complications. Further controlled studies are necessary to validate the role of honey in the management of obesity, both as a preventive and as a therapeutic agent.
Collapse
Affiliation(s)
- Muhammad Faiz Zulkifli
- Cell Signaling and Biotechnology Research Group (CesBTech), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| | - Mohd Naim Fadhli Mohd Radzi
- Cell Signaling and Biotechnology Research Group (CesBTech), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| | - Jonel P. Saludes
- Center for Chemical Biology & Biotechnology (C2B2) and Center for Natural Drug Discovery and Development (CND3), University of San Agustin, Iloilo City, Philippines,Balik Scientist Program, Philippine Council for Health Research and Development, Department of Science and Technology, Taguig, Philippines
| | - Doralyn S. Dalisay
- Center for Chemical Biology & Biotechnology (C2B2) and Center for Natural Drug Discovery and Development (CND3), University of San Agustin, Iloilo City, Philippines,Balik Scientist Program, Philippine Council for Health Research and Development, Department of Science and Technology, Taguig, Philippines
| | - Wan Iryani Wan Ismail
- Cell Signaling and Biotechnology Research Group (CesBTech), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia,Biological Security and Sustainability (BIOSES) Research Group, Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia,Wan Iryani Wan Ismail, Cell Signaling and Biotechnology Research Group (CesBTech), Biological Security and Sustainability (BIOSES) Research Group, Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21300, Kuala Nerus, Terengganu, Malaysia.
| |
Collapse
|
8
|
Depommier C, Everard A, Druart C, Maiter D, Thissen JP, Loumaye A, Hermans MP, Delzenne NM, de Vos WM, Cani PD. Serum metabolite profiling yields insights into health promoting effect of A. muciniphila in human volunteers with a metabolic syndrome. Gut Microbes 2022; 13:1994270. [PMID: 34812127 PMCID: PMC8632301 DOI: 10.1080/19490976.2021.1994270] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Reduction of A. muciniphila relative abundance in the gut microbiota is a widely accepted signature associated with obesity-related metabolic disorders. Using untargeted metabolomics profiling of fasting plasma, our study aimed at identifying metabolic signatures associated with beneficial properties of alive and pasteurized A. muciniphila when administrated to a cohort of insulin-resistant individuals with metabolic syndrome. Our data highlighted either shared or specific alterations in the metabolome according to the form of A. muciniphila administered with respect to a control group. Common responses encompassed modulation of amino acid metabolism, characterized by reduced levels of arginine and alanine, alongside several intermediates of tyrosine, phenylalanine, tryptophan, and glutathione metabolism. The global increase in levels of acylcarnitines together with specific modulation of acetoacetate also suggested induction of ketogenesis through enhanced β-oxidation. Moreover, our data pinpointed some metabolites of interest considering their emergence as substantial compounds pertaining to health and diseases in the more recent literature.
Collapse
Affiliation(s)
- Clara Depommier
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (Welbio), UCLouvain, Université Catholique De Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (Welbio), UCLouvain, Université Catholique De Louvain, Brussels, Belgium
| | - Céline Druart
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (Welbio), UCLouvain, Université Catholique De Louvain, Brussels, Belgium
| | - Dominique Maiter
- Pôle Edin, Institut De Recherches Expérimentales Et Cliniques, UCLouvain, Université Catholique De Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Pôle Edin, Institut De Recherches Expérimentales Et Cliniques, UCLouvain, Université Catholique De Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Audrey Loumaye
- Pôle Edin, Institut De Recherches Expérimentales Et Cliniques, UCLouvain, Université Catholique De Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Michel P. Hermans
- Pôle Edin, Institut De Recherches Expérimentales Et Cliniques, UCLouvain, Université Catholique De Louvain, Brussels, Belgium,Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (Welbio), UCLouvain, Université Catholique De Louvain, Brussels, Belgium
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherland,Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (Welbio), UCLouvain, Université Catholique De Louvain, Brussels, Belgium,CONTACT Patrice D. Cani UCLouvain, Université Catholique De Louvain, Ldri, Metabolism and Nutrition Research Group, Av. E. Mounier, 73 Box B1.73.11, B-1200Brussels, Belgium
| |
Collapse
|
9
|
Untargeted Metabolomics Analysis of the Serum Metabolic Signature of Childhood Obesity. Nutrients 2022; 14:nu14010214. [PMID: 35011090 PMCID: PMC8747180 DOI: 10.3390/nu14010214] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Obesity rates among children are growing rapidly worldwide, placing massive pressure on healthcare systems. Untargeted metabolomics can expand our understanding of the pathogenesis of obesity and elucidate mechanisms related to its symptoms. However, the metabolic signatures of obesity in children have not been thoroughly investigated. Herein, we explored metabolites associated with obesity development in childhood. Untargeted metabolomic profiling was performed on fasting serum samples from 27 obese Caucasian children and adolescents and 15 sex- and age-matched normal-weight children. Three metabolomic assays were combined and yielded 726 unique identified metabolites: gas chromatography–mass spectrometry (GC–MS), hydrophilic interaction liquid chromatography coupled to mass spectrometry (HILIC LC–MS/MS), and lipidomics. Univariate and multivariate analyses showed clear discrimination between the untargeted metabolomes of obese and normal-weight children, with 162 significantly differentially expressed metabolites between groups. Children with obesity had higher concentrations of branch-chained amino acids and various lipid metabolites, including phosphatidylcholines, cholesteryl esters, triglycerides. Thus, an early manifestation of obesity pathogenesis and its metabolic consequences in the serum metabolome are correlated with altered lipid metabolism. Obesity metabolite patterns in the adult population were very similar to the metabolic signature of childhood obesity. Identified metabolites could be potential biomarkers and used to study obesity pathomechanisms.
Collapse
|
10
|
Lan Q, Zhang Y, Lin F, Meng Q, Buys N, Fan H, Sun J. Sex-Specific Associations Between Serum Phosphate Concentration and Cardiometabolic Disease: A Cohort Study on the Community-Based Older Chinese Population. Diabetes Metab Syndr Obes 2022; 15:813-826. [PMID: 35313679 PMCID: PMC8934154 DOI: 10.2147/dmso.s354167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/08/2022] [Indexed: 04/20/2023] Open
Abstract
PURPOSE This study aimed to investigate the association between sex-specific baseline serum phosphate and the incidence of new-onset cardiometabolic disease in a cohort of Shanghai-based older Chinese individuals. PATIENTS AND METHODS A community cohort of 5000 disease-free Chinese men and women was recruited in 2013 and followed until 2017 for the development of cardiometabolic disease. Participants underwent index and follow-up health screens at the Tongji Medical School affiliated Shanghai East Hospital, including blood biochemistry analysis, anthropometric measurements, interview on health-related behaviors, and clinical evaluation. RESULTS Higher baseline serum phosphate (>1.25 mmol/L) was significantly associated with new-onset type-2 diabetes mellitus (HR 1.730, 95% CI 1.127-2.655) and metabolic syndrome (HR 0.640, 95% CI 1.085-2.155) in women. Baseline serum phosphate was associated with age, BMI, waist circumference, SBP, total calcium, bicarbonate, and total cholesterol in women. The estimated risk of developing diabetes mellitus in women with inorganic phosphate >1.25 mmol/L was 14.54%. Inorganic phosphate accounted for 9.2% of the variance explained in a total estimated 14.52% of variance attributed to BMI, total cholesterol, total calcium, waist circumference, and inorganic phosphate. CONCLUSION Serum phosphate concentration showed sex-specific associations with diabetes and metabolic syndrome. Higher inorganic phosphate was associated with increased risk of developing diabetes mellitus in women. These findings may be important in the assessment of individualized metabolic risk.
Collapse
Affiliation(s)
- Qin Lan
- Shanghai East Hospital, Tongji University, Shanghai, People's Republic of China
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yuming Zhang
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD, Australia
| | - Fang Lin
- Shanghai East Hospital, Tongji University, Shanghai, People's Republic of China
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qingshu Meng
- Shanghai East Hospital, Tongji University, Shanghai, People's Republic of China
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Nicholas Buys
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Huimin Fan
- Shanghai East Hospital, Tongji University, Shanghai, People's Republic of China
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Jing Sun
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
11
|
Frangi G, Guicheteau M, Jacquot F, Pyka G, Kerckhofs G, Feyeux M, Veziers J, Guihard P, Halgand B, Sourice S, Guicheux J, Prieur X, Beck L, Beck-Cormier S. PiT2 deficiency prevents increase of bone marrow adipose tissue during skeletal maturation but not in OVX-induced osteoporosis. Front Endocrinol (Lausanne) 2022; 13:921073. [PMID: 36465661 PMCID: PMC9708882 DOI: 10.3389/fendo.2022.921073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
The common cellular origin between bone marrow adipocytes (BMAds) and osteoblasts contributes to the intimate link between bone marrow adipose tissue (BMAT) and skeletal health. An imbalance between the differentiation ability of BMSCs towards one of the two lineages occurs in conditions like aging or osteoporosis, where bone mass is decreased. Recently, we showed that the sodium-phosphate co-transporter PiT2/SLC20A2 is an important determinant for bone mineralization, strength and quality. Since bone mass is reduced in homozygous mutant mice, we investigated in this study whether the BMAT was also affected in PiT2-/- mice by assessing the effect of the absence of PiT2 on BMAT volume between 3 and 16 weeks, as well as in an ovariectomy-induced bone loss model. Here we show that the absence of PiT2 in juveniles leads to an increase in the BMAT that does not originate from an increased adipogenic differentiation of bone marrow stromal cells. We show that although PiT2-/- mice have higher BMAT volume than control PiT2+/+ mice at 3 weeks of age, BMAT volume do not increase from 3 to 16 weeks of age, leading to a lower BMAT volume in 16-week-old PiT2-/- compared to PiT2+/+ mice. In contrast, the absence of PiT2 does not prevent the increase in BMAT volume in a model of ovariectomy-induced bone loss. Our data identify SLC20a2/PiT2 as a novel gene essential for the maintenance of the BMAd pool in adult mice, involving mechanisms of action that remain to be elucidated, but which appear to be independent of the balance between osteoblastic and adipogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Giulia Frangi
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Marie Guicheteau
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Frederic Jacquot
- Nantes Université, CHU Nantes, Inserm, CNRS, CRCI2NA, Nantes, France
| | - Grzegorz Pyka
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UC Louvain, Louvain-la-Neuve, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UC Louvain, Louvain-la-Neuve, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
- IREC, Institute of Experimental and Clinical Research, UC Louvain, Woluwé-Saint-Lambert, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Magalie Feyeux
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Joëlle Veziers
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Pierre Guihard
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Boris Halgand
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Sophie Sourice
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Xavier Prieur
- Nantes Université, CNRS, Inserm, l’Institut du Thorax, Nantes, France
| | - Laurent Beck
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Sarah Beck-Cormier
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
- *Correspondence: Sarah Beck-Cormier,
| |
Collapse
|
12
|
Smita RM, Shuvo APR, Raihan S, Jahan R, Simin FA, Rahman A, Biswas S, Salem L, Sagor MAT. The Role of Mineral Deficiencies in Insulin Resistance and Obesity. Curr Diabetes Rev 2022; 18:e171121197987. [PMID: 34789132 DOI: 10.2174/1573399818666211117104626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/06/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
Minerals are critical for maintaining overall health. These tiny chemical compounds are responsible for enzymatic activation, maintaining healthy teeth and bones, regulating energy metabolism, enhancing immunity, and aiding muscle and brain function. However, mineral deficiency in the form of inadequate or under nourished intake affects millions of people throughout the world, with well-documented adverse health consequences of malnutrition. Conversely, mineral deficiency may also be a risk factor for Insulin Resistance (IR) and obesity. This review focuses on another, more "less discussed" form of malnutrition, namely mineral deficiency and its contribution to metabolic disorders. At the cellular level, minerals maintain not only molecular communication but also trigger several key biochemical pathways. Disturbances in these processes due to mineral insufficiency may gradually lead to metabolic disorders such as insulin resistance, pre-diabetes, and central obesity, which might lead to renal failure, cardiac arrest, hepatic carcinoma, and various neurodegenerative diseases. Here we discuss the burden of disease promoted by mineral deficiencies and the medical, social, and economic consequences. Mineral deficiency-mediated IR and obesity have a considerable negative impact on individual well-being, physical consideration, and economic productivity. We discuss possible molecular mechanisms of mineral deficiency that may lead to IR and obesity and suggest strategies to counter these metabolic disorders. To protect mankind from mineral nutrient deficiencies, the key is to take a variety of foods in reasonable quantities, such as organic and pasture-raised eggs, low fat dairy, and grass-fed and finished meats, insecticide, and pesticide-free vegetables and fruits.
Collapse
Affiliation(s)
| | | | - Sabbir Raihan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Rajib Jahan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Faria Anjum Simin
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Ashiqur Rahman
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Soumick Biswas
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Liyad Salem
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Md Abu Taher Sagor
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
13
|
Cilliers K, Muller CJF. Multi-element Analysis of Brain Regions from South African Cadavers. Biol Trace Elem Res 2021; 199:425-441. [PMID: 32361883 DOI: 10.1007/s12011-020-02158-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022]
Abstract
Trace elements are vital for a variety of functions in the brain. However, an imbalance can result in oxidative stress. It is important to ascertain the normal levels in different brain regions, as such information is still lacking. Therefore, this study aimed to provide baseline trace element concentrations from a South African population, as well as determine trace element differences between sex and brain regions. Samples from the caudate nucleus, putamen, globus pallidus and hippocampus were analysed using inductively coupled plasma mass spectrometry. Aluminium, antimony, arsenic, barium, boron, cadmium, calcium, chromium, cobalt, copper, iron, lead, magnesium, manganese, mercury, molybdenum, nickel, phosphorus, potassium, selenium, silicon, sodium, strontium, vanadium and zinc were assessed. A multiple median regression model was used to determine differences between sex and regions. Twenty-nine male and 13 female cadavers from a Western Cape, South African population were included (mean age 35 years, range 19 to 45). Trace element levels were comparable to those of other populations, although magnesium was considerably lower. While there were no sex differences, significant anatomical regional differences existed; the caudate nucleus and hippocampus were the most similar, and the globus pallidus and hippocampus the most different. In conclusion, this is the first article to report the trace element concentrations of brain regions from a South African population. Low magnesium levels in the brain may be linked to a dietary deficiency, and migraines, depression and epilepsy have been linked to low magnesium levels. Future research should be directed to increase the dietary intake of magnesium.
Collapse
Affiliation(s)
- Karen Cilliers
- Division of Clinical Anatomy, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Western Cape, South Africa.
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Western Cape, South Africa
- Division of Medical Physiology, Faculty of Medicine and Health ScieAnces, Stellenbosch University, Tygerberg, Western Cape, South Africa
| |
Collapse
|
14
|
van Kempen TATG, Deixler E. SARS-CoV-2: influence of phosphate and magnesium, moderated by vitamin D, on energy (ATP) metabolism and on severity of COVID-19. Am J Physiol Endocrinol Metab 2021; 320:E2-E6. [PMID: 33174766 PMCID: PMC7816430 DOI: 10.1152/ajpendo.00474.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/02/2020] [Accepted: 11/08/2020] [Indexed: 01/10/2023]
Abstract
The use of vitamin D to reduce the severity of COVID-19 complications is receiving considerable attention, backed by encouraging data. Its purported mode of action is as an immune modulator. Vitamin D, however, also affects the metabolism of phosphate and Mg, which may well play a critical role in SARS-CoV-2 pathogenesis. SARS-CoV-2 may induce a cytokine storm that drains ATP whose regeneration requires phosphate and Mg. These minerals, however, are often deficient in conditions that predispose people to severe COVID-19, including older age (especially males), diabetes, obesity, and usage of diuretics. Symptoms observed in severe COVID-19 also fit well with those seen in classical hypophosphatemia and hypomagnesemia, such as thrombocytopenia, coagulopathy, dysfunction of liver and kidneys, neurologic disturbances, immunodeficiency, failure of heart and lungs, delayed weaning from a respirator, cardiac arrhythmia, seizures, and, finally, multiorgan failure. Deficiencies of phosphate and Mg can be amplified by kidney problems commonly observed in patients with COVID-19 resulting in their wastage into urine. Available data show that phosphate and Mg are deficient in COVID-19, with phosphate showing a remarkable correlation with its severity. In one experiment, patients with COVID-19 were supplemented with a cocktail of vitamin D3, Mg, and vitamin B12, with very encouraging results. We, thus, argue that patients with COVID-19 should be monitored and treated for phosphate and Mg deficiencies, ideally already in the early phases of infection. Supplementation of phosphate and Mg combined with vitamin D could also be implemented as a preventative strategy in populations at risk.
Collapse
|
15
|
Brener A, Lebenthal Y, Cleper R, Kapusta L, Zeitlin L. Body composition and cardiometabolic health of pediatric patients with X-linked hypophosphatemia (XLH) under burosumab therapy. Ther Adv Endocrinol Metab 2021; 12:20420188211001150. [PMID: 33796255 PMCID: PMC7970173 DOI: 10.1177/20420188211001150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/12/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Burosumab, a recombinant anti-FGF23 monoclonal antibody, was recently introduced as a treatment for X-linked hypophosphatemia (XLH). Burosumab normalizes blood phosphate levels, thereby healing rickets, decreasing leg bowing, and reducing pain. We aimed to explore the body composition and cardiometabolic health of pediatric patients with XLH treated with burosumab. METHODS This observational real-life study was conducted on growing children and adolescents. The outcome measures included changes in sex- and age-adjusted anthropometric and body composition parameters [fat mass (FM), fat-free mass (FFM), appendicular skeletal muscle mass (ASMM), muscle-to-fat ratio (MFR)], blood pressure, laboratory evaluation, and radiographic rickets severity [Thacher Rickets Severity Score (TRSS)]. Body composition was assessed by bioelectrical impedance analysis (BIA). Percentiles for FFM% and ASMM% were calculated according to BIA pediatric reference curves. The delta variable was calculated as the variable at 12 months minus the variable at baseline. RESULTS A total of 15 pediatric patients with XLH are treated in our clinic; included in the analyses were 7 children and adolescents (3 males, mean age 8.7 ± 3.2 years) with XLH without comorbidities. Baseline BIA revealed an unfavorable physique, with increased body fat percentage in five patients and decreased muscle mass in six. Indices of lean body mass significantly increased after 6 and 12 months of treatment: FFM(kg) (p = 0.001, p = 0.046, respectively) and ASMM(kg) (p = 0.012, p = 0.034, respectively), without any significant change in FM(kg). The percentile of ASMM% increased significantly after 6 months of treatment (p = 0.006) and stabilized thereafter. TRSS improved significantly after 12 months of therapy (p = 0.005). Age was positively correlated with delta TRSS (r = 0.814, p = 0.026), and delta TRSS was negatively correlated with delta MFR (r = -0.826, p = 0.022). CONCLUSIONS There was a heretofore unrecognized improvement in body composition of growing children and adolescents with XLH who were treated with burosumab. These findings highlight the need to initiate burosumab treatment at a younger age when rickets is less severe.
Collapse
Affiliation(s)
| | - Yael Lebenthal
- Pediatric Endocrinology and Diabetes Unit,
Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv,
Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv,
Israel
| | - Roxana Cleper
- Pediatric Nephrology Unit, Dana-Dwek Children’s
Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv, Israel
| | - Livia Kapusta
- Pediatric Cardiology Unit, Dana-Dwek Children’s
Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv, Israel
- Department of Paediatric Cardiology, Amalia
Children’s Hospital, Radboud University Medical Centre, Nijmegen, The
Netherlands
| | - Leonid Zeitlin
- Pediatric Orthopedic Department, Dana-Dwek
Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv,
Israel
- Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv, Israel
| |
Collapse
|
16
|
Yang ZY, Kao TW, Peng TC, Chen YY, Yang HF, Wu CJ, Chen WL. Examining the association between serum phosphate levels and leukocyte telomere length. Sci Rep 2020; 10:5438. [PMID: 32214202 PMCID: PMC7096403 DOI: 10.1038/s41598-020-62359-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/10/2020] [Indexed: 12/24/2022] Open
Abstract
Accelerated telomere attrition is related to various diseases, and multiple factors have been reported to influence telomere length. However, little attention has focused on the relationship between serum phosphate levels and mean telomere length. The purpose of this study was to explore the relationship between serum phosphate levels and mean telomere length in the US general population. A total of 7,817 participants from the 1999–2002 NHANES were included. The association between serum phosphate levels and mean telomere length was investigated using regression models. A remarkably positive relationship between serum phosphate levels and mean telomere length emerged after adjustments were made for covariates. The adjusted β coefficient of serum phosphate levels for mean telomere length was 0.038 (95% confidence intervals (CIs), 0.022 to 0.095, p = 0.002). A longer telomere length was observed in participants with serum phosphate levels in the highest quartiles, and a dose-dependent association was observed. Our study demonstrated that higher quartiles of phosphate had a remarkable correlation with longer telomere length.
Collapse
Affiliation(s)
- Zhe-Yu Yang
- Department of General Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tung-Wei Kao
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Clinical Medical, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Tao-Chun Peng
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yuan-Yuei Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Department of Pathology, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Hui-Fang Yang
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chen-Jung Wu
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Family Medicine, Department of Community Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan, Republic of China
| | - Wei-Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China. .,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.
| |
Collapse
|
17
|
Gutiérrez OM, Porter AK, Viggeswarapu M, Roberts JL, Beck GR. Effects of phosphorus and calcium to phosphorus consumption ratio on mineral metabolism and cardiometabolic health. J Nutr Biochem 2020; 80:108374. [PMID: 32278118 DOI: 10.1016/j.jnutbio.2020.108374] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 02/13/2020] [Accepted: 03/04/2020] [Indexed: 12/21/2022]
Abstract
Phosphorus is a common additive used in food processing that is typically consumed in excess of the recommended daily allowance; however, our knowledge of its effects on health, in the context of normal renal function, is limited. Unlike phosphorus, calcium intake is generally less than recommended, and it has been hypothesized that the calcium to phosphorus ratio may be partly responsible for the proposed negative health consequences. Therefore, this study sought to determine the effects of increased phosphorus additive intake, in the context of high calcium consumption, on endocrine markers of mineral metabolism and cardiometabolic health. An outpatient feeding study was performed in which healthy adults were fed a run-in control diet for 2 weeks followed by a phosphorus additive enhanced diet with supplemental calcium to an approximate ratio of 1 (experimental diet) for 2 weeks. Blood and urine samples were collected, and participants had brachial flow-mediated dilatation measured, with analyses comparing follow-up measures to baseline. Two weeks of experimental diet increased serum fibroblast growth factor 23 concentrations but lowered body weight and serum leptin; however, other phosphorus responsive factors such as osteopontin and osteocalcin did not increase. A complementary study in male mice also demonstrated that the regulation of known dietary phosphorus responsive factors was mostly abrogated when dietary calcium was raised in parallel with phosphorus. In conclusion, the study identifies weight, leptin and insulin as responsive to dietary phosphorus and that certain aspects of the systemic phosphorus response are attenuated by a corresponding high calcium intake.
Collapse
Affiliation(s)
- Orlando M Gutiérrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL.
| | - Anna K Porter
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; School of Health Professions, College of Nursing and Health Professions, University of Southern Mississippi, Hattiesburg, MS
| | | | - Joseph L Roberts
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University, Atlanta, GA
| | - George R Beck
- The Atlanta Department of Veterans Affairs Medical Center, Decatur, GA; Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University, Atlanta, GA; The Winship Cancer Institute, Emory University, Atlanta, GA.
| |
Collapse
|
18
|
Obesity and adiposity: the culprit of dietary protein efficacy. Clin Sci (Lond) 2020; 134:389-401. [DOI: 10.1042/cs20190583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/24/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022]
Abstract
AbstractObesity and increased body adiposity have been alarmingly increasing over the past decades and have been linked to a rise in food intake. Many dietary restrictive approaches aiming at reducing weight have resulted in contradictory results. Additionally, some policies to reduce sugar or fat intake were not able to decrease the surge of obesity. This suggests that food intake is controlled by a physiological mechanism and that any behavioural change only leads to a short-term success. Several hypotheses have been postulated, and many of them have been rejected due to some limitations and exceptions. The present review aims at presenting a new theory behind the regulation of energy intake, therefore providing an eye-opening field for energy balance and a potential strategy for obesity management.
Collapse
|
19
|
Zhukouskaya VV, Rothenbuhler A, Colao A, Di Somma C, Kamenický P, Trabado S, Prié D, Audrain C, Barosi A, Kyheng C, Lambert AS, Linglart A. Increased prevalence of overweight and obesity in children with X-linked hypophosphatemia. Endocr Connect 2020; 9:144-153. [PMID: 31910157 PMCID: PMC6993252 DOI: 10.1530/ec-19-0481] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/07/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND/AIM X-linked hypophosphatemia (XLH) is a rare disease characterized by low phosphate levels. Scientific evidence points to a link between hypophosphatemia and obesity in general population. The aim of our longitudinal observational study was to investigate the prevalence of obesity and associated factors in a large cohort of children with XLH. PATIENTS/METHODS We studied 172 XLH-children 5-20 years of age (113 girls/59 boys). Anthropometric parameters (weight, height, and BMI) were collected at birth and during follow-up at mean ages of 5.3, 8.2, 11.3, and 15.9 years (groups 1, 2, 3, and 4, respectively). In each group, subjects were classified based on International Obesity Taskforce (IOTF) cut off values of BMI for age and sex as overweight or obese (IOTF 25-30 or ≥30 kg/m2, respectively). RESULTS In each age-group, almost 1/3 of XLH-patients were classified as overweight or obese (29.4, 28.7, 27.5, and 36.7% in groups 1, 2, 3, and 4, respectively). Children without a XLH-family history had higher BMI-IOTF at every point of follow-up, compared to those with positive XLH-family history. Similarly, higher BMI-IOTF was significantly associated with treatment duration (23.3 ± 4.4 vs 23.8 ± 3.8 vs 25.2 ± 4.5 kg/m2, for subjects with treatment duration of <5, 5-10 and >10 years, respectively, P for trend = 0.025). Multiple regression analysis confirmed an association of treatment duration and lack of XLH-family history with higher BMI-IOTF. CONCLUSION One out of three of XLH-children have phenotypically unfavourable metabolic profile expressed as increased prevalence of overweight or obesity in comparison to general population. Both the lack of XLH family history and the duration of treatment increase the risk of higher BMI-IOTF. BMI should be carefully monitored in children, and later in adults, with XLH.
Collapse
Affiliation(s)
- Volha V Zhukouskaya
- APHP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, FilièreOSCAR and Platform of Expertise for Rare Diseases Paris-Saclay, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
- Division of Endocrinology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
- Correspondence should be addressed to V V Zhukouskaya:
| | - Anya Rothenbuhler
- APHP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, FilièreOSCAR and Platform of Expertise for Rare Diseases Paris-Saclay, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
- APHP, Department of Endocrinology and Diabetology for Children, Bicêtre Paris Saclay Hospital, Le Kremlin-Bicêtre, France
| | - Annamaria Colao
- Division of Endocrinology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carolina Di Somma
- Division of Endocrinology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
- IRCCS SDN, Naples, Italy
| | - Peter Kamenický
- APHP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, FilièreOSCAR and Platform of Expertise for Rare Diseases Paris-Saclay, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
- APHP, Department of Endocrinology and Reproductive Diseases, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
- Paris Sud – Paris Saclay University, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Séverine Trabado
- Paris Sud – Paris Saclay University, Faculté de Médecine, Le Kremlin-Bicêtre, France
- APHP, Department of Molecular Genetics, Pharmacogenetics and Hormonology, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
| | - Dominique Prié
- Université Paris V, Faculté de Médecine, Paris, France
- Hôpital Necker EnfantsMalades APHP, INSERM U1151, Paris, France
| | - Christelle Audrain
- APHP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, FilièreOSCAR and Platform of Expertise for Rare Diseases Paris-Saclay, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
| | - Anna Barosi
- APHP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, FilièreOSCAR and Platform of Expertise for Rare Diseases Paris-Saclay, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
| | - Christèle Kyheng
- APHP, Department of Adolescent Medicine, Bicêtre Paris Saclay Hospital, Le Kremlin-Bicêtre, France
| | - Anne-Sophie Lambert
- APHP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, FilièreOSCAR and Platform of Expertise for Rare Diseases Paris-Saclay, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
- APHP, Department of Endocrinology and Diabetology for Children, Bicêtre Paris Saclay Hospital, Le Kremlin-Bicêtre, France
- APHP, Department of Adolescent Medicine, Bicêtre Paris Saclay Hospital, Le Kremlin-Bicêtre, France
| | - Agnès Linglart
- APHP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, FilièreOSCAR and Platform of Expertise for Rare Diseases Paris-Saclay, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, France
- APHP, Department of Endocrinology and Diabetology for Children, Bicêtre Paris Saclay Hospital, Le Kremlin-Bicêtre, France
- Paris Sud – Paris Saclay University, Faculté de Médecine, Le Kremlin-Bicêtre, France
| |
Collapse
|
20
|
Dandachy S, Mawlawi H, Obeid O. Effect of Processed Chickpea Flour Incorporation on Sensory Properties of Mankoushe Zaatar. Foods 2019; 8:foods8050151. [PMID: 31058863 PMCID: PMC6560399 DOI: 10.3390/foods8050151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Chickpea flour is known to have good nutritional values. Nevertheless, it is commonly made from ground grains, and characterized by an “off-flavor”. Processing of chickpea grains before flour formation reduces the intensity of the off-flavor. Therefore, two experiments were conducted: first to examine the effect of conventional processing (soaking, boiling, and drying) on the nutritional composition of the chickpea flour; and second, to investigate the impact of processed chickpea flour incorporation with different ratios on the sensory properties of mankoushe zaatar, a popular Lebanese pastry, usually made up of refined wheat flour. Chickpea flour was found to be nutritionally superior compared to refined wheat flour, and conventional processing of the flour was found not to affect its content of protein, fats, carbohydrates, and phosphorus, while total dietary and crude fibers were significantly increased. The fatty acid profile was minimally affected, while magnesium and potassium were reduced. The sensory test conducted among panelists (n = 60) showed that the incorporation of processed chickpea flour into the dough of mankoushe zaatar with ratios of 30% and 50% provided an end-product with better taste and overall acceptability compared to the regular mankoushe. Hence, conventionally processed chickpea flour can be used as a fortifier to improve the nutritional quality of bakery products without negatively affecting their sensory properties.
Collapse
Affiliation(s)
- Sahar Dandachy
- Department of Nutrition, Faculty of Public Health, Lebanese University, Tripoli 1300, Lebanon.
| | - Hiba Mawlawi
- Department of Nutrition, Faculty of Public Health, Lebanese University, Tripoli 1300, Lebanon.
- Doctoral School of Science & Technology, Lebanese University, Tripoli 1300, Lebanon.
| | - Omar Obeid
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon.
| |
Collapse
|
21
|
Zohal M, Jam-Ashkezari S, Namiranian N, Moosavi A, Ghadiri-Anari A. Association between selected trace elements and body mass index and waist circumference: A cross sectional study. Diabetes Metab Syndr 2019; 13:1293-1297. [PMID: 31336480 DOI: 10.1016/j.dsx.2019.01.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/17/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND The prevalence of obesity has increased worldwide. Abnormal plasma level of some trace elements may be associated with obesity. The present study was designed to compare the plasma level of zinc, phosphate, calcium and magnesium with the degree of body mass index and waist circumference. MATERIAL AND METHODS In this cross sectional study 149 persons (20-60 years old) from March 2014 till April 2017 were included. Definition of central obesity was waist circumference (WC)≥ 102 cm and ≥88 cm in men and women, respectively. Also BMI categorized to: normal weight: 18.50-24.99, overweight: 25.00-29.99 and obese: ≥30 kg/m2 respectively. Mg, Ca, P and Zinc in plasma was checked after12 h fasting in each persons. Comparison between the level of Mg, P, Ca and Zinc by three categories of BMI or waist circumference performed. The data were analyzed by independent T-test and one-way ANOVA. Scheffe method was used to determine post-hoc pair-wise comparisons. The relationship between BMI and concentration of elements was detected by linear correlation and Cubic model. A p ≤ 0.05 were considered statistically significant. Statistical analyses were executed by SPSS version 20. RESULTS In this study, 32.2% male and remainder female, mean age of 42.26 ± 13.03 were participated. 40.9% were normal and 59.1% obese base on waist circumference. Also 24.8% normal,44.3% overweight and remainder was obese according to BMI. Obese subjects base on waist circumference had significantly lower serum Zinc(pvalue:0.002), Ca (pvalue:0.0001)and Mg(pvalue:0.042) concentration. Whereas, P concentration was significantly higher in obese cases in comparison with normal subjects(pvalue:0.012). Also normal cases had significantly higher serum Zinc (pvalue: 0.0001), Ca (pvalue:0.0001), and Mg(pvalue:0.006) concentration compared to overweight and obese subjects according to BMI categorizes. CONCLUSION Inverse correlation present between plasma zinc, calcium and magnesium level and BMI and waist circumference, but positive correlation seen between P level and waist circumference. Further studies are needed to evaluate the effect of dietary or supplemental interventions on obesity and central obesity.
Collapse
Affiliation(s)
- Mahnaz Zohal
- Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Saeedeh Jam-Ashkezari
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nasim Namiranian
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amin Moosavi
- Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Akram Ghadiri-Anari
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
22
|
Pang C, Yang H, Hu B, Wang S, Chen M, Cohen DS, Chen HS, Jarrell JT, Carpenter KA, Rosin ER, Huang X. Identification and Analysis of Alzheimer's Candidate Genes by an Amplitude Deviation Algorithm. ACTA ACUST UNITED AC 2019; 9. [PMID: 31080696 PMCID: PMC6505709 DOI: 10.4172/2161-0460.1000460] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background: Alzheimer’s disease (AD) is the most common form of senile dementia. However, its pathological mechanisms are not fully understood. In order to comprehend AD pathological mechanisms, researchers employed AD-related DNA microarray data and diverse computational algorithms. More efficient computational algorithms are needed to process DNA microarray data for identifying AD-related candidate genes. Methods: In this paper, we propose a specific algorithm that is based on the following observation: When an acrobat walks along a steel-wire, his/her body must have some swing; if the swing can be controlled, then the acrobat can maintain the body balance. Otherwise, the acrobat will fall. Based on this simple idea, we have designed a simple, yet practical, algorithm termed as the Amplitude Deviation Algorithm (ADA). Deviation, overall deviation, deviation amplitude, and 3δ are introduced to characterize ADA. Results: 52 candidate genes for AD have been identified via ADA. The implications for some of the AD candidate genes in AD pathogenesis have been discussed. Conclusions: Through the analysis of these AD candidate genes, we believe that AD pathogenesis may be related to the abnormality of signal transduction (AGTR1 and PTAFR), the decrease in protein transport capacity (COL5A2 (221729_at), COL5A2 (221730_at), COL4A1), the impairment of axon repair (CNR1), and the intracellular calcium dyshomeostasis (CACNB2, CACNA1E). However, their potential implication for AD pathology should be further validated by wet lab experiments as they were only identified by computation using ADA.
Collapse
Affiliation(s)
- Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Hualan Yang
- College of Mathematics and Software Science, Sichuan Normal University, Chengdu, China
| | - Benqiong Hu
- College of Management Science, Chengdu University of Technology, Chengdu, China
| | - Shipeng Wang
- College of Mathematics and Software Science, Sichuan Normal University, Chengdu, China
| | - Meixia Chen
- College of Mathematics and Software Science, Sichuan Normal University, Chengdu, China
| | - David S Cohen
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hannah S Chen
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Juliet T Jarrell
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kristy A Carpenter
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eric R Rosin
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
23
|
Abdouni L, Olabi A, Obeid O. Postprandial energy expenditure of protein is affected by its phosphorus content. J Therm Biol 2018; 78:214-218. [DOI: 10.1016/j.jtherbio.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/09/2018] [Accepted: 10/13/2018] [Indexed: 10/28/2022]
|
24
|
Imi Y, Yabiki N, Abuduli M, Masuda M, Yamanaka-Okumura H, Taketani Y. High phosphate diet suppresses lipogenesis in white adipose tissue. J Clin Biochem Nutr 2018. [PMID: 30487667 DOI: 10.3164/jcbn.17.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Excessive phosphate intake has been positively associated with renal and vascular dysfunction, conversely negatively associated with body fat accumulation. We investigated the effect of a high-phosphate diet on the expression of lipid metabolic genes in white adipose tissue and liver. Male 8-week-old Sprague-Dawley rats were fed a control diet containing 0.6% phosphate or a high-phosphate diet containing 1.5% phosphate for 4 weeks. In comparison to the control group, the HP group showed a significantly lower body fat mass and fasting plasma insulin level alongside decreased lipogenic and increased lipolytic gene expression in visceral fat. Additionally, the expression of genes involved in hepatic lipogenesis, hepatic glycogenesis, and triglyceride accumulation decreased in the high-phosphate group. Exogenous phosphate, parathyroid hormone, and fibroblast growth factor 23 did not directly affect the expression of lipolytic or lipogenic genes in 3T3-L1 adipocytes and HepG2 hepatocytes. Thus, the high-phosphate diet suppressed the activity of white adipose tissue by increasing lipolytic gene expression and decreasing lipogenic gene expression. These effects could have been caused by the lowered fasting plasma insulin level that occurred in response to the high-phosphate diet, but were not directly caused by the increases in plasma phosphate or phosphaturic hormones.
Collapse
Affiliation(s)
- Yukiko Imi
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Norie Yabiki
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Maerjianghan Abuduli
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
25
|
Haroun D, Mechli R, Sahuri R, AlKhatib S, Obeid O, El Mallah C, Wood L, AlSuwaidi K. Metabolic syndrome among adolescents in Dubai, United Arab Emirates, is attributable to the high prevalence of low HDL levels: a cross-sectional study. BMC Public Health 2018; 18:1284. [PMID: 30463538 PMCID: PMC6249919 DOI: 10.1186/s12889-018-6215-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) describes a combination of risk factors that increase the risk of developing chronic diseases. The prevalences of MetS and its risk factors are increasing, especially in the Arab region. A cross-sectional study was carried out to assess the prevalences of MetS and its associated risk factors among adolescents in the United Arab Emirates (UAE). METHODS A total of 596 students (308 boys and 288 girls) aged 10 to 15.9 years old were recruited from 14 public secondary schools in Dubai, UAE. Anthropometric and biochemical data were measured. RESULTS According to the International Diabetes Federation (IDF) criteria, the prevalence of MetS was 3.7%, and it was more common among boys than girls (12 boys versus 10 girls). MetS was also more likely to be found in students who were obese or overweight than those with normal weight. The most prevalent and significant MetS risk factor was low high-density lipoprotein (HDL) cholesterol levels. CONCLUSIONS This study indicates the importance of carrying out further investigations about the constituents of HDL and their atherogenic effects. Additionally, these results strongly recommend setting a consensus for HDL measurement, since small variations in methodologies may lead to substantial deviations in results.
Collapse
Affiliation(s)
- Dalia Haroun
- Department of Public Health and Nutrition, College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates.
| | - Rola Mechli
- Department of Public Health and Nutrition, College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Razan Sahuri
- Department of Public Health and Nutrition, College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Safa AlKhatib
- Department of Public Health and Nutrition, College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Omar Obeid
- Department of Nutrition and Food Science, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Carla El Mallah
- Department of Nutrition and Food Science, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Lesley Wood
- Department of Public Health and Nutrition, College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Khulood AlSuwaidi
- School Health Center, Ministry of Health and Prevention, Dubai, United Arab Emirates
| |
Collapse
|
26
|
Assaad M, El Mallah C, Obeid O. Phosphorus ingestion with a high-carbohydrate meal increased the postprandial energy expenditure of obese and lean individuals. Nutrition 2018; 57:59-62. [PMID: 30153580 DOI: 10.1016/j.nut.2018.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/16/2018] [Accepted: 05/22/2018] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Phosphorus ingestion with glucose was reported to stimulate the postprandial peripheral uptake of both phosphorus and glucose, a process that favors energy production. The aim of this study was to determine whether phosphorus ingestion with a meal can affect energy metabolism. METHODS Overnight fasted men (eight lean and seven obese) consumed a high-carbohydrate meal (648 kcal) with either placebo or phosphorus (500 mg) tablets in a random order. Energy expenditure and substrate oxidation were monitored for 240 min using ventilated hood indirect calorimetry. RESULTS Phosphorus ingestion with a meal increased the postprandial energy expenditure of both lean and obese individuals (P < 0.001), although in different patterns. Alterations in postprandial substrate oxidation was highly noticeable from time 120 min onward, where phosphorus-treated lean participants exhibited a significant decrease in respiratory quotient. CONCLUSION Phosphorus ingestion with a high-carbohydrate meal alters postprandial energy metabolism mainly by enhancing postprandial energy expenditure that may ultimatly favor weight loss.
Collapse
Affiliation(s)
- Mariam Assaad
- Department of Nutrition and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Carla El Mallah
- Department of Nutrition and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Omar Obeid
- Department of Nutrition and Food Sciences, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
27
|
Imi Y, Yabiki N, Abuduli M, Masuda M, Yamanaka-Okumura H, Taketani Y. High phosphate diet suppresses lipogenesis in white adipose tissue. J Clin Biochem Nutr 2018; 63:181-191. [PMID: 30487667 PMCID: PMC6252294 DOI: 10.3164/jcbn.17-141] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 01/31/2018] [Indexed: 02/06/2023] Open
Abstract
Excessive phosphate intake has been positively associated with renal and vascular dysfunction, conversely negatively associated with body fat accumulation. We investigated the effect of a high-phosphate diet on the expression of lipid metabolic genes in white adipose tissue and liver. Male 8-week-old Sprague-Dawley rats were fed a control diet containing 0.6% phosphate or a high-phosphate diet containing 1.5% phosphate for 4 weeks. In comparison to the control group, the HP group showed a significantly lower body fat mass and fasting plasma insulin level alongside decreased lipogenic and increased lipolytic gene expression in visceral fat. Additionally, the expression of genes involved in hepatic lipogenesis, hepatic glycogenesis, and triglyceride accumulation decreased in the high-phosphate group. Exogenous phosphate, parathyroid hormone, and fibroblast growth factor 23 did not directly affect the expression of lipolytic or lipogenic genes in 3T3-L1 adipocytes and HepG2 hepatocytes. Thus, the high-phosphate diet suppressed the activity of white adipose tissue by increasing lipolytic gene expression and decreasing lipogenic gene expression. These effects could have been caused by the lowered fasting plasma insulin level that occurred in response to the high-phosphate diet, but were not directly caused by the increases in plasma phosphate or phosphaturic hormones.
Collapse
Affiliation(s)
- Yukiko Imi
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Norie Yabiki
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Maerjianghan Abuduli
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
28
|
Cardiac Autonomic Neuropathy as a Result of Mild Hypercaloric Challenge in Absence of Signs of Diabetes: Modulation by Antidiabetic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9389784. [PMID: 29643979 PMCID: PMC5831709 DOI: 10.1155/2018/9389784] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022]
Abstract
Cardiac autonomic neuropathy (CAN) is an early cardiovascular complication of diabetes occurring before metabolic derangement is evident. The cause of CAN remains elusive and cannot be directly linked to hyperglycemia. Recent clinical data report cardioprotective effects of some antidiabetic drugs independent of their hypoglycemic action. Here, we used a rat model receiving limited daily increase in calories from fat (HC diet) to assess whether mild metabolic challenge led to CAN in absence of interfering effects of hyperglycemia, glucose intolerance, or obesity. Rats receiving HC diet for 12 weeks showed reduction in baroreceptor sensitivity and heart rate variability despite lack of change in baseline hemodynamic and cardiovascular structural parameters. Impairment of cardiac autonomic control was accompanied with perivascular adipose inflammation observed as an increased inflammatory cytokine expression, together with increased cardiac oxidative stress, and signaling derangement characteristic of diabetic cardiomyopathy. Two-week treatment with metformin or pioglitazone rectified the autonomic derangement and corrected the molecular changes. Switching rats to normal chow but not to isocaloric amounts of HC for two weeks reversed CAN. As such, we conclude that adipose inflammation due to increased fat intake might underlie development of CAN and, hence, the beneficial effects of metformin and pioglitazone.
Collapse
|
29
|
Miao Z, Feng Y, Zhang J, Tian W, Li J, Yang Y. Regulation of phosphate transport and AMPK signal pathway by lower dietary phosphorus of broilers. Oncotarget 2017; 8:107825-107832. [PMID: 29296204 PMCID: PMC5746106 DOI: 10.18632/oncotarget.22609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/29/2017] [Indexed: 11/25/2022] Open
Abstract
Lower available P (aP) was used as a base value in nutritional strategies for mitigating P pollution by animal excreta. We hypothesized that the mechanism regulating phosphate transport under low dietary P might be related with the AMPK signal pathway. A total of 144 one-day-old Arbor Acres Plus broilers were randomly allocated to control (HP) or trial (LP) diets, containing 0.45 and 0.23% aP, respectively. Growth performance, blood, intestinal, and renal samples were tested in 21-day-old broilers. Results shown that LP decreased body weight gain and feed intake. Higher serum Ca and fructose, but lower serum P and insulin were detected in LP-fed broilers. NaPi-IIb mRNA expression in intestine and NaPi-IIa mRNA expression in kidney were higher in the LP group. AMP: ATP, p-AMPK: total AMPK, and p-ACC: total ACC ratios in the duodenal mucosa were decreased in the LP group, whereas the p-mTOR: total mTOR ratio increased. These findings suggested that the increase in phosphate transport owing to LP diet might be regulated either directly by higher mTOR activity or indirectly by the suppressive AMPK signal, with corresponding changes in blood insulin and fructose content. A novel viewpoint on the regulatory mechanism underlying phosphate transport under low dietary P conditions was revealed, which might provide theoretical guidelines for reducing P pollution by means of nutritional regulation.
Collapse
Affiliation(s)
- Zhiqiang Miao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Yan Feng
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Junzhen Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Wenxia Tian
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jianhui Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Yu Yang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| |
Collapse
|
30
|
Phosphorus Supplementation Recovers the Blunted Diet-Induced Thermogenesis of Overweight and Obese Adults: A Pilot Study. Nutrients 2016; 8:nu8120801. [PMID: 27941661 PMCID: PMC5188456 DOI: 10.3390/nu8120801] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/27/2016] [Accepted: 12/06/2016] [Indexed: 11/17/2022] Open
Abstract
Diet-induced thermogenesis (DIT) is believed to be largely related to ATP production, which is dependent on phosphorus (P) availability. We aimed to test the effect of P addition on DIT of lean and overweight/obese healthy subjects. DIT was measured with or without P in 10 lean and 13 overweight/obese adults in a double-blind randomized cross-over pilot study with one week washout period. After 10 h overnight fast, resting metabolic rate, respiratory quotient, and substrate utilization were measured at fasting and every 30 min for 3 h after subjects drank a standardized glucose solution, with P (500 mg) or placebo pills. Subjective ratings of hunger and satiety were assessed before and after the end of each experiment using validated visual analogue scale (VAS) questionnaires. Overweight/obese subjects had a blunted DIT with placebo, while P supplementation induced a 23% increase in their DIT area under the curve (p < 0.05), which was associated with a significant increase in carbohydrate oxidation. Subjects had lower appetite following P supplementation, which was expressed as a significantly (p = 0.02) lower desire to eat a meal (4.0 ± 0.7 cm) compared with placebo (5.8 ± 0.9 cm). P supplementation recovers the blunted diet-induced thermogenesis in overweight and obese subjects and enhances their postprandial satiety.
Collapse
|
31
|
Park B, Kim J. Oral Contraceptive Use, Micronutrient Deficiency, and Obesity among Premenopausal Females in Korea: The Necessity of Dietary Supplements and Food Intake Improvement. PLoS One 2016; 11:e0158177. [PMID: 27348598 PMCID: PMC4922824 DOI: 10.1371/journal.pone.0158177] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/10/2016] [Indexed: 01/01/2023] Open
Abstract
This study addressed the associations between oral contraceptive (OC) use and obesity as measured by recording the body mass index (BMI) of premenopausal females, and possible interactions with micronutrient intake were considered. A group of 39,189 premenopausal females aged 35-59 were included in the analysis; they were in the Health Examinee cohort. Participant BMIs were calculated from anthropometric measurements, and females with a BMI≥25kg/m2 were considered obese. Individual OC use, age at first OC use, duration of OC use, nutrient intake, and other covariates were measured with a structured questionnaire. A multivariate logistic regression with an interaction term was applied to identify the odds ratio (OR) and 95% confidence intervals (CI) between OC use and obesity along with consideration of micronutrient intake interactions. OC use is associated with an increased risk of obesity (OR = 1.12, 95% CI = 1.04-1.20), and females who used OCs for more than 6 months over their lifetimes were more likely to be obese (OR = 1.15, 95% CI = 1.01-1.32) compared with those who used OCs for <6 months. There were interaction effects between phosphorus, potassium, vitamin A, vitamin B1, vitamin B2, niacin, vitamin C intake and total duration of OC use on being obesity (P-value<0.05). When stratified by micronutrient intake, the associations between total OC use duration and obesity were present only among those with calcium, phosphorus, potassium, vitamin A, B1, B2, C, niacin, and folate intakes below the recommended levels. Efforts to estimate nutrient intake and prevent micronutrient depletion with supplements or food should be considered by clinicians for females who take OC for a long period.
Collapse
Affiliation(s)
- Boyoung Park
- Department of Cancer Control and Policy, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Korea
- Cancer Early Detection Branch, Division of Cancer Management Policy, National Cancer Control Institute, National Cancer Center, Goyang-si, Korea
| | - Jeongseon Kim
- Department of Cancer Control and Policy, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Korea
- Molecular Epidemiology Branch, Division of Cancer Epidemiology and Prevention, Research Institute, National Cancer Center, Goyang-si, Korea
| |
Collapse
|
32
|
Effect of phosphorus supplementation on weight gain and waist circumference of overweight/obese adults: a randomized clinical trial. Nutr Diabetes 2015; 5:e189. [PMID: 26690287 PMCID: PMC4735052 DOI: 10.1038/nutd.2015.38] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/19/2015] [Accepted: 11/10/2015] [Indexed: 11/25/2022] Open
Abstract
Background: Phosphorus status is inversely correlated with body weight; however, the effect of phosphorus supplementation on body weight in a controlled design has not been studied. Methods: This is a double-blind, randomized, placebo-controlled trial of 63 adults aged 18–45 years with a body mass index (BMI) of ⩾25 kg m−2 and normal kidney function at the American University of Beirut. Participants were randomly assigned to the placebo or phosphorus group where daily placebo or phosphorus supplements were ingested with three main meals (breakfast, lunch and dinner) for a period of 12 weeks. Primary outcomes were changes in anthropometric measures, blood metabolites (including lipid profile, glucose and insulin) and subjective appetite scores. The trial is registered with Clinical Trial.gov, NCT02329990. Results: Body weight was significantly lower in the phosphorus group when compared with the placebo group (−0.65 kg (95% confidence interval (CI) −1.69 to 0.40) vs 1.13 kg (95% CI 0.19 to 2.06), P=0.01). Similarly, BMI and waist circumference were significantly lower in the phosphorus group when compared with the placebo group (−0.24 kg m−2 (95% CI −0.59 to 0.12) vs 0.42 kg m−2 (95% CI 0.05 to 0.78), P=0.01; −3.62 cm (95% CI−4.90 to −2.33) vs 0.38 cm ( 95% CI−0.44 to 1.20), P<0.001; respectively). Several parameters of subjective appetite scores were decreased in the phosphorus-supplemented group. Conclusions: Phosphorus supplementation for 12 weeks significantly decreases body weight, BMI, waist circumference and subjective appetite scores. These findings support a promising role of the mineral phosphorus in the prevention and management of obesity, especially abdominal adiposity. The exact mechanisms of action and longer-term effects still need to be elucidated.
Collapse
|
33
|
Phosphorus ingestion improves oral glucose tolerance of healthy male subjects: a crossover experiment. Nutr J 2015; 14:112. [PMID: 26514124 PMCID: PMC4627612 DOI: 10.1186/s12937-015-0101-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 10/12/2015] [Indexed: 12/25/2022] Open
Abstract
Background Fasting serum phosphorus (P) was reported to be inversely related to serum glucose and insulin, while the impact of P ingestion is not well documented. The effect of P intake with or before glucose ingestion on postprandial glucose and insulin statuses was investigated. Method Two cross over experiments using healthy male subjects were conducted. Experiment 1: Overnight fasted subjects (n = 7) randomly received: 500 mg of P tablets, glucose (75 g) solution with placebo or 500 mg of P tablets. Experiment 2: Overnight fasted subjects (n = 8) underwent similar procedures to those of experiment 1, except that placebo or 500 mg P tablets were given 60 min prior to glucose ingestion. Results In both experiments, serum P decreased following glucose ingestion. Co-ingestion of P with glucose improved, at time 60 min, postprandial glucose (P < 0.05), insulin (P < 0.05), and insulin sensitivity index (p < 0.006), while P pre-ingestion failed to exert similar effect. Conclusion This study suggests that postprandial glucose and insulin are affected by exogenous P supply, especially when co-ingested with glucose.
Collapse
|
34
|
Huffman JE, Albrecht E, Teumer A, Mangino M, Kapur K, Johnson T, Kutalik Z, Pirastu N, Pistis G, Lopez LM, Haller T, Salo P, Goel A, Li M, Tanaka T, Dehghan A, Ruggiero D, Malerba G, Smith AV, Nolte IM, Portas L, Phipps-Green A, Boteva L, Navarro P, Johansson A, Hicks AA, Polasek O, Esko T, Peden JF, Harris SE, Murgia F, Wild SH, Tenesa A, Tin A, Mihailov E, Grotevendt A, Gislason GK, Coresh J, D'Adamo P, Ulivi S, Vollenweider P, Waeber G, Campbell S, Kolcic I, Fisher K, Viigimaa M, Metter JE, Masciullo C, Trabetti E, Bombieri C, Sorice R, Döring A, Reischl E, Strauch K, Hofman A, Uitterlinden AG, Waldenberger M, Wichmann HE, Davies G, Gow AJ, Dalbeth N, Stamp L, Smit JH, Kirin M, Nagaraja R, Nauck M, Schurmann C, Budde K, Farrington SM, Theodoratou E, Jula A, Salomaa V, Sala C, Hengstenberg C, Burnier M, Mägi R, Klopp N, Kloiber S, Schipf S, Ripatti S, Cabras S, Soranzo N, Homuth G, Nutile T, Munroe PB, Hastie N, Campbell H, Rudan I, Cabrera C, Haley C, Franco OH, Merriman TR, Gudnason V, Pirastu M, Penninx BW, Snieder H, Metspalu A, Ciullo M, Pramstaller PP, van Duijn CM, Ferrucci L, Gambaro G, Deary IJ, Dunlop MG, Wilson JF, Gasparini P, Gyllensten U, Spector TD, Wright AF, Hayward C, Watkins H, Perola M, Bochud M, Kao WHL, Caulfield M, Toniolo D, Völzke H, Gieger C, Köttgen A, Vitart V. Modulation of genetic associations with serum urate levels by body-mass-index in humans. PLoS One 2015; 10:e0119752. [PMID: 25811787 PMCID: PMC4374966 DOI: 10.1371/journal.pone.0119752] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 02/03/2015] [Indexed: 11/17/2022] Open
Abstract
We tested for interactions between body mass index (BMI) and common genetic variants affecting serum urate levels, genome-wide, in up to 42569 participants. Both stratified genome-wide association (GWAS) analyses, in lean, overweight and obese individuals, and regression-type analyses in a non BMI-stratified overall sample were performed. The former did not uncover any novel locus with a major main effect, but supported modulation of effects for some known and potentially new urate loci. The latter highlighted a SNP at RBFOX3 reaching genome-wide significant level (effect size 0.014, 95% CI 0.008-0.02, Pinter= 2.6 x 10-8). Two top loci in interaction term analyses, RBFOX3 and ERO1LB-EDARADD, also displayed suggestive differences in main effect size between the lean and obese strata. All top ranking loci for urate effect differences between BMI categories were novel and most had small magnitude but opposite direction effects between strata. They include the locus RBMS1-TANK (men, Pdifflean-overweight= 4.7 x 10-8), a region that has been associated with several obesity related traits, and TSPYL5 (men, Pdifflean-overweight= 9.1 x 10-8), regulating adipocytes-produced estradiol. The top-ranking known urate loci was ABCG2, the strongest known gout risk locus, with an effect halved in obese compared to lean men (Pdifflean-obese= 2 x 10-4). Finally, pathway analysis suggested a role for N-glycan biosynthesis as a prominent urate-associated pathway in the lean stratum. These results illustrate a potentially powerful way to monitor changes occurring in obesogenic environment.
Collapse
Affiliation(s)
- Jennifer E Huffman
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Massimo Mangino
- King's College London, St. Thomas' Hospital Campus, London, United Kingdom
| | - Karen Kapur
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Toby Johnson
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nicola Pirastu
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy; University of Trieste, Trieste, Italy
| | - Giorgio Pistis
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Lorna M Lopez
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Perttu Salo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Anuj Goel
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Toshiko Tanaka
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Abbas Dehghan
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Giovanni Malerba
- Biology and Genetics section, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Albert V Smith
- Icelandic Heart Association Research Institute, Kopavogur, Iceland; University of Iceland, Reykjavik, Iceland
| | - Ilja M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Laura Portas
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | | | - Lora Boteva
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Pau Navarro
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Asa Johansson
- Uppsala Clinical Research Center, Uppsala University Hospital, Upsalla, Sweden; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, 751 85, Sweden
| | - Andrew A Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy; Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Croatia, Soltanska 2, Split, 21000, Croatia
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia; Broad Institute, Cambridge, MA, United States of America; Children's Hospital Boston, Boston, MA, United States of America
| | - John F Peden
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom; Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | - Federico Murgia
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Sarah H Wild
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Albert Tenesa
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom; Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | | | - Anne Grotevendt
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Gauti K Gislason
- Icelandic Heart Association Research Institute, Kopavogur, Iceland
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America; Welch Center for Prevention, Epidemiology and Clinical Research, John Hopkins University, Baltimore, MD, United States of America
| | - Pio D'Adamo
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy; University of Trieste, Trieste, Italy
| | - Sheila Ulivi
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Gerard Waeber
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Susan Campbell
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Ivana Kolcic
- Faculty of Medicine, University of Split, Croatia, Soltanska 2, Split, 21000, Croatia
| | - Krista Fisher
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Margus Viigimaa
- Tallinn University of Technology, Department of Biomedical Engineering, Chair of Medical Physics, Tallinn, Estonia; Centre of Cardiology, North Estonia Medical Centre, Tallinn, Estonia
| | - Jeffrey E Metter
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Corrado Masciullo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Elisabetta Trabetti
- Biology and Genetics section, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Cristina Bombieri
- Biology and Genetics section, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Rossella Sorice
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Angela Döring
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Eva Reischl
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | - Albert Hofman
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Melanie Waldenberger
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - H-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-University, Munich, Germany; Klinikum Grosshadern, Munich, Germany
| | - Gail Davies
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alan J Gow
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola Dalbeth
- Bone and Joint Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Lisa Stamp
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Johannes H Smit
- Department of Psychiatry/EMGO Institute, VU University Medical Centre, Amsterdam, the Netherlands
| | - Mirna Kirin
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Ramaiah Nagaraja
- Laboratory of Genetics, National Institute on Aging (NIA), Baltimore, MD, United States of America
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Claudia Schurmann
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Kathrin Budde
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Susan M Farrington
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Evropi Theodoratou
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Antti Jula
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Turku, Finland
| | - Veikko Salomaa
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | | | - Michel Burnier
- Department of Medicine, Nephrology Division, Lausanne University Hospital, Lausanne, Switzerland
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Norman Klopp
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Sabine Schipf
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Samuli Ripatti
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Turku, Finland; Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom; University of Helsinki, Institute of Molecular Medicine, Helsinki, Finland
| | - Stefano Cabras
- Department of Mathematics and Informatics, Università di Cagliari, Cagliari, Italy; Department of Statistics, Universidad Carlos III de Madrid, Madrid, Spain
| | - Nicole Soranzo
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Teresa Nutile
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nicholas Hastie
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Harry Campbell
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Igor Rudan
- Faculty of Medicine, University of Split, Croatia, Soltanska 2, Split, 21000, Croatia; Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | | | - Chris Haley
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom; Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Oscar H Franco
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Vilmundur Gudnason
- Icelandic Heart Association Research Institute, Kopavogur, Iceland; University of Iceland, Reykjavik, Iceland
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Brenda W Penninx
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands; Department of Epidemiology, Subdivision Genetic Epidemiology, Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Marina Ciullo
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Peter P Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy; Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Cornelia M van Duijn
- Department of Epidemiology, Subdivision Genetic Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Giovanni Gambaro
- Institute of Internal Medicine, Renal Program, Columbus-Gemelli University Hospital, Catholic University, Rome, Italy
| | - Ian J Deary
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Malcolm G Dunlop
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - James F Wilson
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Paolo Gasparini
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy; University of Trieste, Trieste, Italy
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, 751 85, Sweden
| | - Tim D Spector
- King's College London, St. Thomas' Hospital Campus, London, United Kingdom
| | - Alan F Wright
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Hugh Watkins
- on behalf of PROCARDIS; Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Markus Perola
- Estonian Genome Center, University of Tartu, Tartu, Estonia; Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Helsinki, Finland; University of Helsinki, Institute of Molecular Medicine, Helsinki, Finland
| | - Murielle Bochud
- University Institute of Social and Preventive Medicine, Lausanne, Switzerland
| | - W H Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America; Welch Center for Prevention, Epidemiology and Clinical Research, John Hopkins University, Baltimore, MD, United States of America
| | - Mark Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America; Renal Division, Freiburg University Hospital, Freiburg, Germany
| | - Veronique Vitart
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
35
|
Abstract
Antiobesity effects of bamboo salt (BS) were evaluated compared with those of purified salt and solar salt by oral administration in a diet-induced obesity model using C57BL/6 mice. Compared with other salts, BS, especially nine times baked BS (BS-9×), significantly reduced body weight, food efficiency ratio, and weights of epididymal adipose tissue and liver in high-fat diet-fed mice. Furthermore, BS suppressed the expression of adipogenic factors, such as CCAAT/enhancer binding protein alpha (C/EBPα), peroxisome proliferator-activated receptor gamma (PPARγ), and sterol regulatory element-binding protein 1c (SREBP-1c). Therefore, BS may suppress obesity by downregulating adipogenesis.
Collapse
Affiliation(s)
- Jaehyun Ju
- Department of Food Science and Nutrition, Pusan National University, Busan, South Korea
| | - Jia-Le Song
- Department of Food Science and Nutrition, Pusan National University, Busan, South Korea
| | - Kun-Young Park
- Department of Food Science and Nutrition, Pusan National University, Busan, South Korea
| |
Collapse
|
36
|
Obeid OA, Hachem DH, Ayoub JJ. Refeeding and metabolic syndromes: two sides of the same coin. Nutr Diabetes 2014; 4:e120. [PMID: 24979149 PMCID: PMC4079929 DOI: 10.1038/nutd.2014.21] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/09/2014] [Accepted: 05/28/2014] [Indexed: 12/13/2022] Open
Abstract
Refeeding syndrome describes the metabolic and clinical changes attributed to aggressive rehabilitation of malnourished subjects. The metabolic changes of refeeding are related to hypophosphatemia, hypokalemia, hypomagnesemia, sodium retention and hyperglycemia, and these are believed to be mainly the result of increased insulin secretion following high carbohydrate intake. In the past few decades, increased consumption of processed food (refined cereals, oils, sugar and sweeteners, and so on) lowered the intake of several macrominerals (mainly phosphorus, potassium and magnesium). This seems to have compromised the postprandial status of these macrominerals, in a manner that mimics low grade refeeding syndrome status. At the pathophysiological level, this condition favored the development of the different components of the metabolic syndrome. Thus, it is reasonable to postulate that metabolic syndrome is the result of long term exposure to a mild refeeding syndrome.
Collapse
Affiliation(s)
- O A Obeid
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences. American University of Beirut, Beirut, Lebanon
| | - D H Hachem
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences. American University of Beirut, Beirut, Lebanon
| | - J J Ayoub
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences. American University of Beirut, Beirut, Lebanon
| |
Collapse
|