scholarly journals Coffee consumption and plasma biomarkers of metabolic and inflammatory pathways in US health professionals

2019 ◽  
Vol 109 (3) ◽  
pp. 635-647 ◽  
Author(s):  
Dong Hang ◽  
Ane Sørlie Kværner ◽  
Wenjie Ma ◽  
Yang Hu ◽  
Fred K Tabung ◽  
...  

ABSTRACT Background Coffee consumption has been linked to lower risk of various health outcomes. However, the biological pathways mediating the associations remain poorly understood. Objectives The aim of this study was to assess the association between coffee consumption and concentrations of plasma biomarkers in key metabolic and inflammatory pathways underlying common chronic diseases. Methods We investigated the associations of total, caffeinated, and decaffeinated coffee consumption with 14 plasma biomarkers, including C-peptide, insulin-like growth factor 1 (IGF-1), IGF binding protein (IGFBP) 1, IGFBP-3, estrone, total and free estradiol, total and free testosterone, sex hormone–binding globulin (SHBG), total adiponectin, high-molecular-weight (HMW) adiponectin, leptin, C-reactive protein (CRP), interleukin 6 (IL-6), and soluble tumor necrosis factor receptor 2 (sTNFR-2). Data were derived from 2 cohorts of 15,551 women (Nurses’ Health Study) and 7397 men (Health Professionals Follow-Up Study), who provided detailed dietary data before blood draw and were free of diabetes, cardiovascular disease, or cancer at the time of blood draw. Multivariable linear regression was used to calculate the percentage difference of biomarker concentrations comparing coffee drinkers with nondrinkers, after adjusting for a variety of demographic, clinical, and lifestyle factors. Results Compared with nondrinkers, participants who drank ≥4 cups of total coffee/d had lower concentrations of C-peptide (−8.7%), IGFBP-3 (−2.2%), estrone (−6.4%), total estradiol (−5.7%), free estradiol (−8.1%), leptin (−6.4%), CRP (−16.6%), IL-6 (−8.1%), and sTNFR-2 (−5.8%) and higher concentrations of SHBG (5.0%), total testosterone (7.3% in women and 5.3% in men), total adiponectin (9.3%), and HMW adiponectin (17.2%). The results were largely similar for caffeinated and decaffeinated coffee. Conclusion Our data indicate that coffee consumption is associated with favorable profiles of numerous biomarkers in key metabolic and inflammatory pathways. This trial was registered at clinicaltrials.gov as NCT03419455.

Author(s):  
Jongeun Rhee ◽  
Erikka Loftfield ◽  
Neal D Freedman ◽  
Linda M Liao ◽  
Rashmi Sinha ◽  
...  

Abstract Background Coffee consumption has been associated with a reduced risk of some cancers, but the evidence for renal cell carcinoma (RCC) is inconclusive. We investigated the relationship between coffee and RCC within a large cohort. Methods Coffee intake was assessed at baseline in the National Institutes of Health–American Association of Retired Persons Diet and Health Study. Among 420 118 participants eligible for analysis, 2674 incident cases were identified. We fitted Cox-regression models to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for coffee consumption vs non-drinkers. Results We observed HRs of 0.94 (95% CI 0.81, 1.09), 0.94 (0.81, 1.09), 0.80 (0.70, 0.92) and 0.77 (0.66, 0.90) for usual coffee intake of <1, 1, 2–3 and ≥4 cups/day, respectively (Ptrend = 0.00003). This relationship was observed among never-smokers (≥4 cups/day: HR 0.62, 95% CI 0.46, 0.83; Ptrend = 0.000003) but not ever-smokers (HR 0.85, 95% CI 0.70, 1.05; Ptrend = 0.35; Pinteraction = 0.0009) and remained in analyses restricted to cases diagnosed >10 years after baseline (HR 0.65, 95% CI 0.51, 0.82; Ptrend = 0.0005). Associations were similar between subgroups who drank predominately caffeinated or decaffeinated coffee (Pinteraction = 0.74). Conclusion In this investigation of coffee and RCC, to our knowledge the largest to date, we observed a 20% reduced risk for intake of ≥2 cups/day vs not drinking. Our findings add RCC to the growing list of cancers for which coffee consumption may be protective.


2016 ◽  
Vol 8 (3) ◽  
pp. 19
Author(s):  
Choaping Ng ◽  
Felicity J Rose ◽  
Sahar Keshvari ◽  
Marina M Reeves ◽  
Goce Dimeski ◽  
...  

<p>Adiponectin is a beneficial adipocyte-secreted hormone, which circulates in a variety of multimeric forms termed low and high molecular weight (LMW/HMW). Effectiveness of clinical therapeutic trials which target adiponectin rely on accurate determination of circulating total and HMW adiponectin levels but the accuracy may be influenced by variations in sample handling processes. The aim of this pilot study was to investigate the effects of delayed processing of blood samples on the concentration of total and HMW adiponectin.</p><p>Materials and Methods: Fasting blood samples were collected for analysis of total and HMW adiponectin concentrations in EDTA plasma and serum from eight healthy participants.  Samples were centrifuged post 15 min storage at 4<sup>o</sup>C as the comparative ‘ideal’ method or after up to 72 h of refrigerated storage or 6 h at room temperature. Total and HMW adiponectin concentrations were measured by ELISA.</p><p>Results: Under ideal handling conditions measurements of total and HMW adiponectin concentrations were significantly higher in serum than in plasma (mean difference: -1.3 µg/mL [95% CI: -1.6, -1.0], p&lt;0.001; and, -0.6 µg/mL [95% CI: -0.7, -0.5], p&lt;0.001, respectively).  Storage of blood samples at 4<sup>o</sup>C for 72 h resulted in significant reductions in concentration of total adiponectin in serum (mean difference: -1.4 µg/mL [95% CI: -2.0, -0.8], p=0.001) and HMW adiponectin in plasma (mean difference: -0.6 µg/mL [95%CI: -0.9, -0.2], p=0.007), compared with ideal conditions.  Further analysis of serum samples showed a significant decrease in total adiponectin concentration after 6 h storage at 4<sup>o</sup>C (mean difference: -1.4 µg/mL [95% CI: -2.0, -0.8], p=0.001) compared with ideal conditions.</p><p>Conclusions: Delayed processing of samples may have differential effects on the concentration of total and HMW adiponectin in serum or plasma. Larger studies are warranted for clinical intervention trials.</p>


2020 ◽  
Author(s):  
Dong Hang ◽  
Oana A. Zeleznik ◽  
Xiaosheng He ◽  
Marta Guasch-Ferre ◽  
Xia Jiang ◽  
...  

<b>Objective:</b> Coffee may protect against multiple chronic diseases, particularly type 2 diabetes, but the mechanisms remain unclear. <p><b>Research Design and Methods: </b>Leveraging dietary and metabolomic data in two large cohorts of women (the Nurses’ Health Study I and II), we identified and validated plasma metabolites associated with coffee intake in 1595 women. We then evaluated the prospective association of coffee-related metabolites with diabetes risk and the added predictivity of these metabolites for diabetes in two nested case-control studies (n=457 cases and 1371 controls). </p> <p><b>Results: </b>Of 461 metabolites, 34 were identified and validated to be associated with total coffee intake, including 13 positive associations (primarily trigonelline, polyphenol metabolites, and caffeine metabolites) and 21 inverse associations (primarily triacylglycerols and diacylglycerols). These associations were generally consistent for caffeinated and decaffeinated coffee, except for caffeine and its metabolites that were only associated with caffeinated coffee intake. The three cholesteryl esters positively associated with coffee intake showed inverse associations with diabetes risk, whereas the 12 metabolites negatively associated with coffee (five diacylglycerols and seven triacylglycerols) showed positive associations with diabetes. Adding the 15 diabetes-associated metabolites to classical risk factors-based prediction model increased the C-statistic from 0.79 (95% CI: 0.76, 0.83) to 0.83 (95% CI: 0.80, 0.86) (<i>P</i><0.001). Similar improvement was observed in the validation set.</p> <p><b>Conclusion: </b>Coffee consumption is associated with widespread metabolic changes, among which lipid metabolites may be critical for the anti- diabetes benefit of coffee. Coffee-related metabolites might help improve prediction of diabetes, but further validation studies <a>are </a>needed.</p>


2020 ◽  
Vol 4 (9) ◽  
Author(s):  
Rachel M Cole ◽  
Sarah Puchala ◽  
Jia-Yu Ke ◽  
Mahmoud Abdel-Rasoul ◽  
Kristin Harlow ◽  
...  

ABSTRACT Background The onset of menopause increases the risk of metabolic syndrome (MetS). Adiponectin is an adipokine associated with insulin sensitivity that is lower in people with MetS. Supplementing diets with linoleic acid (LA)-rich oil increased adiponectin concentrations and improved glucose control in women with type 2 diabetes. The effect of LA on adipokines, especially total and the bioactive form of adiponectin, high-molecular-weight (HMW) adiponectin, in women with MetS is unknown. Objectives The aim of this study was to explore the effect of supplementation of the diet with an oil rich in LA on adipokines in women with MetS. The effect of the LA-rich oil (LA-oil) on oxylipins, key metabolites that may influence inflammation and metabolism, was also explored. Methods In this open-label single-arm pilot study, 18 postmenopausal nondiabetic women with MetS enrolled in a 2-phase study were instructed to consume LA-rich vegetable oil (10 mL/d) as part of their habitual diets. Women consumed an oleic acid–rich oil (OA-oil) for 4 wk followed by an LA-oil for 16 wk. Fasting concentrations of adipokines, fatty acids, oxylipins, and markers of glycemia and inflammation were measured. Results After 4 wk of OA-oil consumption, fasting glucose and total adiponectin concentrations decreased whereas fasting C-reactive protein increased. After 16 wk of LA-oil supplementation total and HMW adiponectin and plasma oxylipins increased. Markers of inflammation and glycemia were unchanged after LA-oil consumption. Conclusions Supplementation with LA-oil increased total and HMW adiponectin concentrations and altered plasma oxylipin profiles. Larger studies are needed to elucidate the links between these changes and MetS. This trial was registered at clinicaltrials.gov as NCT02063165.


2020 ◽  
Vol 4 (Supplement_2) ◽  
pp. 639-639
Author(s):  
Bryant Keirns ◽  
Destinee Elliott ◽  
Christina Sciarrillo ◽  
Nicholas Koemel ◽  
Kara Poindexter ◽  
...  

Abstract Objectives Common clinical recommendations direct patients to report fasted when blood work (e.g., triglycerides [TG], glucose [Glu]) will take place, which typically excludes black coffee consumption. Despite its negligible calorie content, caffeinated coffee increases fatty acid mobilization. However, whether this effect meaningfully alters fasting metabolic testing or influences the results of a fat tolerance test is unclear. We investigated whether allowing black coffee intake within a fast prior to blood work affected fasting TG and Glu, as well as the postprandial lipemic and glycemic response following an abbreviated fat tolerance test. Methods In a randomized crossover design, participants were instructed to consume only water, or were allowed 8 fluid ounces of black coffee at the end of a 10-hr fast. Next, TG and Glu were assessed using the Cholestech LDX system (Alere Cholestech: Hayward, CA, USA) before and after a previously validated 4-hr fat tolerance test (9 kcal/kg; 73% fat, 26% CHO). Paired t-tests were performed to assess baseline and 4-hr values, absolute change, and % change for both TG and Glu. Results Preliminary analysis of healthy subjects (n = 3 of 10 subjects completed; 1 M/2F; age 20.3 ± 2.3; BMI 25.7 ± 0.6) revealed that consuming coffee prior to the blood draw did not affect fasting TG (Mean difference (MD) = 7.0 mg/dL; P = 0.68). Similarly, the lipemic response was not altered by coffee, evidenced by no alterations in 4-hr TG (MD = 7.6 mg/dL), Δ TG (MD = 14.7 mg/dL), and % change in TG (MD = 29.1%; all P’s ≥ 0.52). Fasting Glu was unchanged following coffee consumption (MD = 29.1 mg/dL; P = 0.90), and indicators of the glycemic response such as 4-hr Glu (MD = 0.0 mg/dL), Δ Glu (MD = 1.0 mg/dL), and % change (MD = 1.2%), were similar among water and coffee trials (all P's ≥ 0.73). Conclusions At this point in the study, coffee consumption does not seem to alter fasting TG or markers of fat tolerance. Additionally, fasting Glu and the glycemic response do not appear to be influenced by coffee consumption. When completed, this study will help answer the practical question of whether coffee need be avoided prior to basic metabolic testing or a fat tolerance test, which may provide increased consistency in metabolic assessment and potentially improve patients’ clinical experience. Funding Sources Lew Wentz Research Scholars Program at Oklahoma State University.


2017 ◽  
Vol 465 ◽  
pp. 30-33 ◽  
Author(s):  
Merel van Andel ◽  
Madeleine L. Drent ◽  
Antonius E. van Herwaarden ◽  
Mariëtte T. Ackermans ◽  
Annemieke C. Heijboer

2007 ◽  
Vol 85 (8) ◽  
pp. 823-830 ◽  
Author(s):  
J. Shearer ◽  
E.A. Sellars ◽  
A. Farah ◽  
T.E. Graham ◽  
D.H. Wasserman

Epidemiological studies indicate that regular coffee consumption reduces the risk of developing type 2 diabetes. Despite these findings, the biological mechanisms by which coffee consumption exerts these effects are unknown. The aim of this study was twofold: to develop a rat model that would further delineate the effects of regular coffee consumption on glucose kinetics, and to determine whether coffee, with or without caffeine, alters the actions of insulin on glucose kinetics in vivo. Male Sprague–Dawley rats were fed a high-fat diet for 4 weeks in combination with one of the following: (i) drinking water as placebo (PL), (ii) decaffeinated coffee (2 g/100 mL) (DC), or (iii) alkaloid caffeine (20 mg/100 mL) added to decaffeinated coffee (2 g/100 mL) (CAF). Catheters were chronically implanted in a carotid artery and jugular vein for sampling and infusions, respectively. Recovered animals (5 days postoperative) were fasted for 5 h before hyperinsulinemic-euglycemic clamps (2 mU·kg–1·min–1). Glucose was clamped at 6 mmol/L and isotopes (2-deoxy-[14C]glucose and [3-3H]glucose) were administered to obtain indices of whole-body and tissue-specific glucose kinetics. Glucose infusion rates and measures of whole-body metabolic clearance were greater in DC than in PL or CAF, indicating increased whole-body insulin sensitivity. As the only difference between DC and CAF was the addition of alkaloid caffeine, it can be concluded that caffeine antagonizes the beneficial effects of DC. Given these findings, decaffeinated coffee may represent a nutritional means of combating insulin resistance.


2012 ◽  
Vol 50 (4) ◽  
Author(s):  
Takuya Imatoh ◽  
Motonobu Miyazaki ◽  
Ken Kadowaki ◽  
Shinichi Tanihara ◽  
Chinami Akashi ◽  
...  

AbstractIn many studies, high-molecular-weight (HMW) adiponectin has been considered the active form of adiponectin. However, whether HMW adiponectin is a good surrogate marker for coronary artery disease still needs to be elucidated.We conducted a hospital-based cross-sectional study to examine the relationship between total, HMW or non-HMW adiponectin concentrations and coronary stenosis in 83 male patients and 138 male controls.Patients with coronary stenosis had significantly lower total adiponectin concentrations compared with controls. Non-HMW adiponectin concentrations in cases were significantly lower than the controls. However, there were no significant differences between cases and controls in HMW adiponectin concentrations. From the receiver operating characteristic (ROC) analysis, the area under the curve (AUC) for total and non-HMW adiponectin was significantly larger than that for HMW adiponectin concentrations. Of the three models, that for non-HMW adiponectin showed the largest AUC (total adiponectin 0.74, HMW adiponectin 0.54, and non-HMW adiponectin 0.79).Despite associations between total adiponectin levels and coronary stenosis, our data go against any apparent association between HMW adiponectin concentrations and coronary stenosis.


2020 ◽  
Vol 124 (8) ◽  
pp. 785-796
Author(s):  
Tommy H. T. Wong ◽  
Jennifer M. F. Wan ◽  
Iris M. Y. Tse ◽  
W. H. Sit ◽  
Jimmy C. Y. Louie

AbstractThe present study aimed to compare the effects of drinking different types of coffee before a high-glycaemic index (GI) meal on postprandial glucose metabolism and to assess the effects of adding milk and sugar into coffee. In this randomised, crossover, acute feeding study, apparently healthy adults (n 21) consumed the test drink followed by a high-GI meal in each session. Different types of coffee (espresso, instant, boiled and decaffeinated, all with milk and sugar) and plain water were tested in separate sessions, while a subset of the participants (n 10) completed extra sessions using black coffees. Postprandial levels of glucose, insulin, active glucagon-like peptide 1 (GLP-1) and nitrotyrosine between different test drinks were compared using linear mixed models. Results showed that only preloading decaffeinated coffee with milk and sugar led to significantly lower glucose incremental AUC (iAUC; 14 % lower, P = 0·001) than water. Preloading black coffees led to greater postprandial glucose iAUC than preloading coffees with milk and sugar added (12–35 % smaller, P < 0·05 for all coffee types). Active GLP-1 and nitrotyrosine levels were not significantly different between test drinks. To conclude, preloading decaffeinated coffee with milk and sugar led to a blunted postprandial glycaemic response after a subsequent high-GI meal, while adding milk and sugar into coffee could mitigate the impairment effect of black coffee towards postprandial glucose responses. These findings may partly explain the positive effects of coffee consumption on glucose metabolism.


Sign in / Sign up

Export Citation Format

Share Document