Skip to main content
Erschienen in: Nutrition & Metabolism 1/2011

Open Access 01.12.2011 | Research

Acute and second-meal effects of almond form in impaired glucose tolerant adults: a randomized crossover trial

verfasst von: Alisa M Mori, Robert V Considine, Richard D Mattes

Erschienen in: Nutrition & Metabolism | Ausgabe 1/2011

Abstract

Background

Nut consumption may reduce the risk of developing type 2 diabetes. The aim of the current study was to measure the acute and second-meal effects of morning almond consumption and determine the contribution of different nut fractions.

Methods

Fourteen impaired glucose tolerant (IGT) adults participated in a randomized, 5-arm, crossover design study where whole almonds (WA), almond butter (AB), defatted almond flour (AF), almond oil (AO) or no almonds (vehicle - V) were incorporated into a 75 g available carbohydrate-matched breakfast meal. Postprandial concentrations of blood glucose, insulin, non-esterified free fatty acids (NEFA), glucagon-like peptide-1 (GLP-1) and appetitive sensations were assessed after treatment breakfasts and a standard lunch.

Results

WA significantly attenuated second-meal and daylong blood glucose incremental area under the curve (AUCI) and provided the greatest daylong feeling of fullness. AB and AO decreased blood glucose AUCI in the morning period and daylong blood glucose AUCI was attenuated with AO. WA and AO elicited a greater second-meal insulin response, particularly in the early postprandial phase, and concurrently suppressed the second-meal NEFA response. GLP-1 concentrations did not vary significantly between treatments.

Conclusions

Inclusion of almonds in the breakfast meal decreased blood glucose concentrations and increased satiety both acutely and after a second-meal in adults with IGT. The lipid component of almonds is likely responsible for the immediate post-ingestive response, although it cannot explain the differential second-meal response to AB versus WA and AO.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1743-7075-8-6) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests. The funding body did not participate in the study design, data collection, analysis and interpretation of data, writing of the manuscript, or in the decision to submit findings for publication.

Authors' contributions

AM and RM participated in the conception of the study. AM, RM, and RC participated in study design and data interpretation. AM participated in conduction of the experiment. All authors read and approved the final manuscript.

Background

The 2025 worldwide projection of IGT is 418 million (8.1% of the adult population) [1]. Lifestyle modification, including nutrition is the cornerstone of its management. Macro- and micronutrients, fiber content, and other components of the diet modulate meal-induced insulin secretion through changes in gastrointestinal transit time and nutrient absorption rates. Additionally, the content of one meal has the potential to affect insulin sensitivity at a second meal by altering circulating NEFA concentrations and daylong insulin demands [2].
Almonds are a low-glycemic index (GI) food, with high fiber, unsaturated fat and low carbohydrate content. There is an inverse relationship between nut consumption and risk of developing type 2 diabetes [3]. In addition, almond consumption increases satiety, reduces cardiovascular disease risk, decreases postprandial glycemia and moderates oxidative damage [4]. The component(s) of almonds responsible for these effects have not been determined. Almonds contain phytates and phenolics, that confer antioxidant, anti-inflammatory and lipid-lowering properties and inhibit trypsin and amylase activity [5]. A decreased rate of nutrient digestion may explain reported increases in satiety and blunted blood glucose response with almond consumption. Stimulation of the incretin and ileal-brake hormone, GLP-1, may also contribute. Consequently, almond consumption may be an effective dietary management tool in insulin resistant individuals who would benefit from replacement of saturated fat with unsaturated fat [6].
Inclusion of 60 g of almonds in meals of healthy individuals decreases glycemia, insulinemia, and postprandial oxidative damage as measured by increased protein thiol concentration [7]. Adding almonds (30-90 g) to a high-GI meal results in a dose-responsive decrease in 2-hour postprandial blood glucose AUCI [8]. However, consumption of almond oil with defatted almond flour, to mimic a bioaccessible almond form, significantly decreased 3-hour blood glucose AUCI with no difference in insulin response compared to when small almond particles were consumed [9]. Similarly, increased and sustained concentrations of cholecystokinin (CCK) and augmented hunger were reported with bioavailable almond oil compared to whole almonds [10]. This suggests the bioavailability of the lipid fraction may be responsible for decreased postprandial glycemia.
The present study evaluated the effects of whole almonds, almond oil, defatted almond flour, and almond butter on acute and second-meal postprandial blood glucose, insulin, NEFA, and GLP-1 concentrations, as well as satiety sensations, in IGT adults.

Methods

Eligibility criteria included: age 18-60 years; not taking medications known to affect glycemia, sleep, or appetite; weight stable (3 month fluctuation of <5 kg); regular breakfast consumer (≥100 kcal ingested within 2 hours of waking on ≥4 d/wk) o blood donation in the previous 3 months; no nut or relevant food allergy; at least one of the following risk factors: A) self-reported family history of type 2 diabetes; B) blood pressure ≥130/85 mmHg; C) fasting blood glucose >5.6 mmol/l; or D) waist circumference (men ≥102 cm; women ≥88 cm); and a 2-hour blood glucose value of 7.8 and <11.1 mmol/l (i.e., IGT) [11]. Height, weight, and body composition were measured using a wall-mounted stadiometer, a clinical scale, and bioelectrical impedence, respectively. A 2-hour, 75-gram oral glucose tolerance test (OGTT) was conducted at a second visit with participants in an 8-10 hour fasted state. The research was approved by the University Institutional Review Board.
One hundred-seventy individuals completed the first screening visit, of which 132 were eligible for and completed the second screening visit. Fourteen participants met all screening criteria and completed the full study protocol. Calculation of power indicated that 13 individuals were necessary to detect a change in blood glucose of 0.35 mmol/l (α = 0.05; Power = 0.80, SD = 0.3)[12]. Participant characteristics are shown in Table 1.
Table 1
Participant characteristics¹
Age (y)
39.3 ± 10.9
Weight (kg)
92.6 ± 19.3
BMI (kg/m²)
33.0 ± 6.9
Body fat (%)
35.8 ± 14.0
Waist circumference (cm)
105.3 ± 16.3
Systolic blood pressure (mmHg)
130.1 ± 11.1
Diastolic blood pressure (mmHg)
82.0 ± 10.0
Fasting blood glucose (mmol/l)
5.5 ± 0.5
Fasting serum insulin (pmol/l)
88.8 ± 46.2
Total cholesterol (mmol/l)
5.42 ± 1.14
HDL-C (mmol/l)
1.18 ± 0.45
LDL-C (mmol/l)
3.16 ± 1.07
Cholesterol:HDL-C ratio
5.1 ± 1.8
Triglycerides (mmol/l)
2.80 ± 1.37
Blood glucose after 2-hour OGTT (mmol/l)
8.3 ± 0.3
QUICKI²
0. 330 ± 0.039
1N = 14 (8 M, 6 F); Mean ± SD
2Quantitative insulin sensitivity check index = 1/[log (fasting insulin, μU/mL) + log (fasting glucose, mg/dL)] [14]
The study utilized a randomized, 5-arm, crossover, single-blinded design. Overnight fasted (8-10 hours) participants reported to the laboratory on 5 occasions separated by at least one week. Menstruating female participants completed test days within the follicular phase of their menstrual cycle. Individuals were requested to maintain their normal exercise, eating, and sleeping patterns and, if taking vitamins or medications, to take them at the same time every day before reporting for testing. Participants were also requested to consume the same meal each evening before reporting to the laboratory at their customary breakfast time.
Upon arrival to the laboratory, participants were weighed and body composition was determined. An indwelling catheter was placed and a baseline blood sample collected. Appetite ratings were scored on a 100 mm visual analogue scale (VAS) anchored with descriptors of "not at all" and "extremely." Next, the participant consumed a test breakfast within 10 minutes that consisted of orange juice and farina [prepared Cream of Wheat®, B&G Foods, Inc.] containing: nothing V (vehicle), whole almonds (WA), almond butter (AB), defatted almond flour (AF) or almond oil (AO) in randomized order. Almonds were non-pareil, dry-roasted and added to the farina whole [provided by the Almond Board of California (Modesto, CA)]. Almonds and their processed forms were from the same almond harvest. Test breakfasts were matched on available carbohydrate and sweetness (nutrient composition shown in Table 2). The amount of almonds added to the cereal was 42.5 grams (~33 almonds) in accord with the Food and Drug Administration (FDA) qualified health claim regarding daily nut intake [13]. After completion of the breakfast meal, palatability of the foods was rated on a VAS (mean palatability scores are shown in Table 3).
Table 2
Test breakfast and lunch nutrient composition
 
Vehicle
Whole
Almond
Almond
Almond
Lunch
  
Almond
Butter
Flour
Oil
 
Energy (kcal)
347
580
580
377
537
374
Weight (g)
644.0
683.5
674.9
656.9
665.5
393.6
Fat (g)
1
22.6
22.6
1
22.6
1.6
Protein (g)
7
16
16
16
7
11.4
Dietary fiber (g)
2.1
7.1
7.1
3.6
2.1
2.8
Soluble fiber (g)
1.4
1.9
1.9
1.5
1.4
1.3
Insoluble fiber (g)
0.7
5.2
5.2
2.1
0.7
1.5
Available carbohydrate (g)
75
75
75
75
75
75
Table 3
Mean palatability scores for test foods¹
 
Vehicle
Whole
Almond
Almond
Almond
  
Almond
Butter
Flour
Oil
Cereal
0.56 ± 0.06a
0.61 ± 0.07a
0.57 ± 0.07a
0.52 ± 0.08a, b
0.37 ± 0.08b
Orange Juice
0.75 ± 0.05
0.78 ± 0.05
0.73 ± 0.06
0.77 ± 0.04
0.73 ± 0.05
Bagel
0.59 ± 0.06
0.64 ± 0.06
0.58 ± 0.04
0.66 ± 0.05
0.65 ± 0.05
¹ Different superscripts indicate significant difference within the meal period (P < 0.05); values represent the score on a 100 mm visual analog scale.
Blood was drawn and appetite was rated 15, 45, 60, 90, 120, 180, and 240 minutes after completion of the test breakfast. At minute 240, participants consumed a standard lunch within 10 minutes that consisted of a plain white bagel, grape or strawberry jelly, and tap water (250 ml). Palatability of the lunch was rated on a VAS. Afternoon blood sampling and appetite scoring occurred using the same time intervals as the morning.
Three milliliters of blood were collected in a red top vacutainer at each draw. After clotting and centrifugation, serum was aliquoted and stored at -80°C for later analysis of insulin, glucose, and NEFA. Four ml of blood were collected in ice-cooled EDTA-plasma vacutainer, 40 μl DPP-IV inhibiter (Millipore, St. Charles, MO) was added, and samples were handled according to manufacturer's directions for analysis of GLP-1. All samples were analyzed in duplicate. Insulin and glucose were measured by electrochemiluminescence and the hexokinase method, respectively. Sensitivity of the insulin immunoassay was 1.39 pmol/l (within-run coefficient of variation (CV) of 1.9%). Hexokinase sensitivity was 0.12 mmol/l (within-run CV of 0.41%). NEFA were analyzed with an enzymatic colorimetric method on an automated analyzer with a sensitivity of 0.00014 mEq/L (within-run CV of 0.75%). GLP-1 was assessed by radioimmunoassay. Sensitivity of the assay was 3 pmol/l (within-run CV of 30.3%). Lipid panel assessment was conducted by MidAmerica Clinical Laboratories, Indianapolis, IN.
Nutrient data were analyzed with the Nutrition Data Systems for Research 2008 (University of Minnesota, Minneapolis, MN). Statistical testing was conducted with SPSS, Version 17.0 (SPSS Inc., Chicago, IL). Repeated measures analysis of variance (ANOVA) was used to explore main effects and, when appropriate, post hoc analyses were conducted with Bonferroni adjustment. Significance was set at p < 0.05. Data are represented as Mean ± SEM. Area under the Curve (AUC) with respect to increase (I) was computed to measure concentration change over time [14] (Formula 1) and quantitative insulin sensitivity check index (QUICKI) was calculated [15].
Fasting concentrations served as baseline for daylong (0-490 minutes) and morning responses (time 0-240 minutes) and the blood sample taken at 240 minutes served as the baseline for afternoon responses (time 240-490 minutes).
Formula 1. Incremental Area Under the Curve
A U C I = ( i = 1 n 1 ( m i + m ( i + 1 ) ) t 1 2 ) ( m 1 i = 1 n 1 t i )
Where n equals the total amount of measurements, mi equals the individual measurements, and ti equals the time between measurements [14].

Results

Blood glucose

Fasting blood glucose concentrations were similar across treatment arms. Mean morning glucose concentrations were greater after consumption of V as compared to WA, AB, and AO (P < 0.02). Afternoon response was greater with AB than all others (P < 0.002). Mean daylong blood glucose change from baseline was lower after consumption of WA compared to AB, AF and V (P < 0.02) (Figure 1C). Daylong blood glucose response was lower with WA compared to V (P < 0.05).
V resulted in a greater blood glucose peak than AB and AO (P < 0.001) with a trend in comparison with WA (P = 0.069). The post-lunch peak was greater with AB compared to WA, AF, and V (P < 0.02) and approached significance compared to AO (P = 0.055).
AUCI was less in WA compared to AB, AF, and V and greater in AB versus AO (P < 0.05) (Figure 2). Morning AUCI with AO was lower than AF and V (P < 0.01) and WA was lower than V (P < 0.04). Afternoon AUCI was lower after WA compared to AB and AO (P < 0.04).

Serum Insulin and Insulin Sensitivity

Fasting insulin was greater in WA compared to V and AF; AO was greater than AF and AB (P < 0.05). The change of insulin from baseline in the morning and over the test day did not differ between treatments. The afternoon insulin response to V was less than WA, AB, and AO, whereas both AO and WA were greater than AF (P < 0.04) (Figure 1B). No differences in absolute peak concentrations occurred in the morning, but the afternoon peak was greater with WA compared to AF and AO compared to AF and V (P < 0.02).
AUCI was greater in AB versus V (P < 0.03) (Figure 2). Postprandial breakfast AUCI for AB was greater than V (P < 0.04). WA postprandial lunch AUCI was greater than AF and V (P < 0.05), and AF was less than AO (P < 0.01).
WA QUICKI was less than that for AB and AF (P < 0.05) and trended lower compared to V (P = 0.068). AO QUICKI was less than AB and AF (P < 0.02).

Nefa

Fasting NEFA concentrations with AF were lower than AB and V (P < 0.02) and approached significance compared to AO (P = 0.06). Morning NEFA concentrations were greater with AB than all others (P < 0.05). AB was greater than V in the afternoon (P < 0.01). Daylong concentrations were lower with AF compared to both AB and AO (P < 0.03) and V was less than AB (P < 0.001) (Figure 1A). Absolute peak response in both time periods was greater in AB than in AF (P < 0.05). WA morning peak concentration tended to be lower than AB (P = 0.053).
Morning and daylong NEFA AUCI were less suppressed in AF versus V (P < 0.03) (Figure 2). NEFA AUCI following WA trended toward greater suppression than V in the morning and over the day (P = 0.067). Afternoon AUCI was less suppressed in AB versus WA and AO (P < 0.05).

Plasma GLP-1

Fasting GLP-1 concentrations were similar between visits (19.99 ± 1.28 pmol/l). There were no significant treatment effects.

Appetite Ratings

No differences in fasting or treatment-related hunger ratings occurred. Daylong fullness ratings were significantly higher in WA compared to AF, AO, and V (P < 0.04) and in AB compared to AF and V (P < 0.01) (Figure 3). Fullness was greater in the morning with WA versus AF and AO and in AB versus AF (P < 0.04), with WA trending higher than V (P = 0.058). Afternoon fullness ratings were greater in WA compared to AF and V (P < 0.001), lower in AF versus AB (P < 0.01) and lower in V compared to AB and AO (P < 0.03).

Palatability

Mean palatability was lower for the farina containing AO compared to WA, AB, and V (P < 0.04), but no sample rated in the lower third of the hedonic scale (Table 3).

Body Weight and Composition

There was no difference in body weight, BMI, or any measures of body composition across treatment visits.

Discussion

The aims of this study included confirmation that nut consumption improves the metabolic profile with respect to diabetes risk; determination of the relative contributions of different almond fractions on these indices and whether acute post-prandial benefits translate to improved insulin sensitivity at a subsequent eating event (second-meal effect). To enhance the ecological validity of the work, whole almonds were included to explore effects with natural mastication and the quantity of almonds included in the test meal corresponded to the recommended intake level in the FDA approved qualified health claim for nuts [13].
In the current study, significantly greater fasting insulin concentrations with WA and AO led to lower QUICKI values and therefore less calculated insulin sensitivity. Due to its correlation with the hyperinsulinemic euglycemic clamp [15], QUICKI is the preferred method for quantifying insulin sensitivity in populations with perturbed insulin sensitivity. Despite higher baseline insulin concentrations, consumption of WA and AO decreased morning blood glucose AUCI compared to V. Postprandial breakfast insulin and NEFA AUCI were not greater after consumption of WA and AO suggestive of greater insulin sensitivity (e.g. the decreased blood glucose AUCI was not determined by a concurrent increased insulin response). Similarly, consumption of 60 g of almonds with white bread decreased 2-hour blood glucose and insulin AUCI in healthy individuals compared to a control meal [7]. Moreover, in healthy men, bioaccessible almond composition produced a lower 3-hour blood glucose response with no significant difference in the insulin or NEFA response [9]. Larger almond particles did not produce the same effect. Previous data from our laboratory did not find a clear relationship between amount of almond chewing (predefined number of chews) and changes in glucose and insulin concentrations in a group of healthy participants [16]. Discrepancies between studies may be due to differences in almond particle sizes (e.g., naturally masticated versus predefined) which could alter nutrient bioaccessibility.
In contrast to lipid-containing treatments, the treatments with little fat (V and AF) produced the largest immediate postprandial glucose responses. The role of fat in decelerating gastric emptying may be partly responsible [17]. Although the AF treatment contained polyphenolic compounds, there was no evidence of impairment of starch digestion in the current study as has been previously reported [18]. NEFA concentrations after consumption of AF were lower than V in the morning postprandial period without differences in insulin concentrations, indicating a slight improvement in NEFA suppression. In comparison, no difference in NEFA concentrations between the combination of AF and AO, large almond particles, and control sunflower oil [9] suggests minimal benefit to the presence of the defatted flour fraction on metabolic risk outcomes.
One study suggested the NEFA concentration 4 hours after a test breakfast accounted for ~50% of the variability in the glycemic response to a standard lunch [19]. We found no significant difference at this time point and do not confirm that NEFA concentrations explain second-meal metabolic differences. However, AB resulted in the lowest overall degree of NEFA suppression in the morning period and was associated with the greatest blood glucose response to the standard lunch. The overall NEFA response in the period before the meal may be a stronger determinant of the second-meal glycemic response than the concentration immediately preceding the second meal. Additionally, no differences were observed in glucose, insulin, or GLP-1 concentrations at 4 hours after the test breakfast, suggesting other determinants of second-meal effects. While the mechanism remains uncertain, this trial confirms the phenomenon. Prior work revealed that inclusion of slowly digestible carbohydrate (e.g., lentils) in a breakfast meal reduced the glucose response after lunch [20]. We show that inclusion of a high unsaturated fat load with breakfast is also effective. Together, these data support the efficacy of dietary moderation of carbohydrate absorption kinetics from a morning meal for extended glycemic control in populations at risk for or with type 2 diabetes.
The high unsaturated fatty acid composition of almonds may account for the blunted glucose concentrations noted in the postprandial period. Acute consumption of PUFA and MUFA decreases postprandial glucose AUCI without altering insulin concentrations [21] due to increased efficiency of insulin receptor signaling and increased glycemic control through stimulation of GLP-1 [6]. Although no significant treatment effects were detected in GLP-1 concentrations, WA and AO led to an overall greater and sustained GLP-1 response that may have contributed to blunted second-meal blood glucose concentrations [22] and modified satiety [23].
The role of almond lipid bioavailability in triggering the release of gut peptides and contributing to energy balance is complicated by differences in the metabolic profiles following WA and AO versus AB consumption. Previous research in healthy participants showed lower breakfast and increased afternoon blood glucose AUCI after consumption of a standard lunch when peanut butter or butter was consumed in a mixed breakfast meal [24]. Our data show this similar afternoon rebound with consumption of AB in the breakfast meal, which cannot be attributed
solely to the lipid component. Additionally, in vitro gastric and duodenal digestion modeling found greater duodenal lipid digestion in finely ground almond particles compared to defatted finely ground almonds with almond oil added back, suggesting that differential dispersion of the lipid (e.g. different surface areas of the lipid droplets) may determine bioaccessibility [25]. Altering the physical form of nuts may have unexpected metabolic effects that warrant further investigation.
Differences in fullness were not likely due to variations in the macronutrient content of the test foods. All provided 75 g of available carbohydrate and the treatments matched on protein, fat, total and soluble fiber, and energy led to variable satiety responses. The cognitive influence of the visual cue of WA and the increased orosensory stimulation from chewing may be responsible for satiety effects [26]. Lipid consumed in oil form previously induced a greater and sustained CCK response and greater satiety in women versus consumption of WA [10], a finding not confirmed in the current study.
An unavoidable limitation of the current study was that breakfast meals were not matched on energy, volume, or macronutrient composition. Due to the study design, available carbohydrate (the main determinant of GI) was standardized between all treatments and macronutrients were matched when possible. The subjective palatability of the treatments was not consistent, with AO considered significantly less desirable than all other treatments except for AF. However, post hoc covariate analysis did not reveal palatability significantly altered results. Additionally, participants were instructed to consume the same meal before reporting for each visit, although significant differences were found. Fewer calories were consumed the night before WA compared to V (~200 kcal) and a lower percent of calories from carbohydrate was consumed the night before AO compared to AB (~5%). The extent to which these differences can explain postprandial breakfast responses is unknown. Greatly altering the GI of a dinner may produce a varied response after a breakfast meal, but the absorptive characteristics between the evening meals consumed before test days in the current study were unlikely so drastically different as those that have previously been shown to produce carry-over effects to the breakfast meal [27]. The possibility exists that differences in dietary intake may also be an artifact of the difficulty of accurately assessing dietary intake and single meal reporting precludes the ability to employ calculations such as the Goldberg cut-off to determine plausibility of reported intake. Nonetheless, the macronutrient and energy intake data appear to fall within normal ranges (49-54% energy from carbohydrate, 28-33% energy from fat, 18-19% energy from protein, and ~30% of mean estimated daily energy requirements).
In summary, inclusion of almonds in the breakfast meal of IGT adults decreased blood glucose concentrations and increased satiety acutely and after a second meal. The lipid component of the almond appears to be largely responsible for the immediate post-ingestive response, although it cannot account for the second-meal response. Overall, daylong glucose, insulin and NEFA concentrations were attenuated in the WA and AO treatments, indicating an improved hormonal profile with their consumption. Importantly, the absolute magnitude of the blood glucose-lowering response equals that achieved with acute administration of acarbose in individuals with IGT [28], suggesting the physiological relevance and applicability of the current findings.

Acknowledgements

We thank the Almond Board of California for funding the project and for providing the almond test foods. We thank the assistance of William Horn for development and adaptation of the Appetite Log VAS software (US Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA 95616). Additional thanks goes to those individuals who participated in the study, to Jenna Potts for assistance with determination of GLP-1 concentrations, to Robert Pazdro for assistance with NEFA determinations, and to Robin Rhine, Judy George, and Tammy Summers for phlebotomy.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests. The funding body did not participate in the study design, data collection, analysis and interpretation of data, writing of the manuscript, or in the decision to submit findings for publication.

Authors' contributions

AM and RM participated in the conception of the study. AM, RM, and RC participated in study design and data interpretation. AM participated in conduction of the experiment. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
2.
Zurück zum Zitat Wong JMW, Jenkins DJA: Carbohydrate digestibility and metabolic effects. J Nutr. 2007, 137: 2539S-2546S. Wong JMW, Jenkins DJA: Carbohydrate digestibility and metabolic effects. J Nutr. 2007, 137: 2539S-2546S.
3.
Zurück zum Zitat Jiang R, Manson JE, Stampfer MJ, Liu S, Willett WC, Hu FB: Nut and peanut butter consumption and risk of type 2 diabetes in women. J Am Med Assoc. 2002, 288: 2554-2560. 10.1001/jama.288.20.2554.CrossRef Jiang R, Manson JE, Stampfer MJ, Liu S, Willett WC, Hu FB: Nut and peanut butter consumption and risk of type 2 diabetes in women. J Am Med Assoc. 2002, 288: 2554-2560. 10.1001/jama.288.20.2554.CrossRef
4.
Zurück zum Zitat Ahrens S, Venkatachalam M, Mistry AM, Lapsley K, Sathe SK: Almond (Prunus dulcis L.) protein quality. Plant Foods for Hum Nutr. 2005, 60: 123-128. 10.1007/s11130-005-6840-2.CrossRef Ahrens S, Venkatachalam M, Mistry AM, Lapsley K, Sathe SK: Almond (Prunus dulcis L.) protein quality. Plant Foods for Hum Nutr. 2005, 60: 123-128. 10.1007/s11130-005-6840-2.CrossRef
5.
Zurück zum Zitat Garrido I, Monagas M, Gómez-Cordovés C, Bartolomé B: Polyphenols and antioxidant properties of almond skins: influence of industrial processing. J Food Sci. 2008, 73: C106-C115. 10.1111/j.1750-3841.2007.00637.x.CrossRef Garrido I, Monagas M, Gómez-Cordovés C, Bartolomé B: Polyphenols and antioxidant properties of almond skins: influence of industrial processing. J Food Sci. 2008, 73: C106-C115. 10.1111/j.1750-3841.2007.00637.x.CrossRef
6.
Zurück zum Zitat Risérus U, Willett WC, Hu FB: Dietary fats and prevention of type 2 diabetes. Prog Lipid Res. 2009, 48: 44-51.CrossRef Risérus U, Willett WC, Hu FB: Dietary fats and prevention of type 2 diabetes. Prog Lipid Res. 2009, 48: 44-51.CrossRef
7.
Zurück zum Zitat Jenkins DJA, Kendall CWC, Josse AR: Almonds decrease postprandial glycemia, insulinemia, and oxidative damage in healthy individuals. J Nutr. 2006, 136: 2987-2992. Jenkins DJA, Kendall CWC, Josse AR: Almonds decrease postprandial glycemia, insulinemia, and oxidative damage in healthy individuals. J Nutr. 2006, 136: 2987-2992.
8.
Zurück zum Zitat Josse AR, Kendall CWC, Augustin LSA, Ellis PR, Jenkins DJA: Almonds and postprandial glycemia--a dose-response study. Met Clin Exp. 2007, 56: 400-404.CrossRef Josse AR, Kendall CWC, Augustin LSA, Ellis PR, Jenkins DJA: Almonds and postprandial glycemia--a dose-response study. Met Clin Exp. 2007, 56: 400-404.CrossRef
9.
Zurück zum Zitat Berry SEE, Tydeman EA, Lewis HB: Manipulation of lipid bioaccessibility of almond seeds influences postprandial lipemia in healthy human subjects. Am J Clin Nutr. 2008, 88: 922-929. Berry SEE, Tydeman EA, Lewis HB: Manipulation of lipid bioaccessibility of almond seeds influences postprandial lipemia in healthy human subjects. Am J Clin Nutr. 2008, 88: 922-929.
10.
Zurück zum Zitat Burton-Freeman B, Davis PA, Schneeman BO: Interaction of fat availability and sex on postprandial satiety and cholecystokinin after mixed-food meals. Am J Clin Nutr. 2004, 80: 1207-1214. Burton-Freeman B, Davis PA, Schneeman BO: Interaction of fat availability and sex on postprandial satiety and cholecystokinin after mixed-food meals. Am J Clin Nutr. 2004, 80: 1207-1214.
11.
Zurück zum Zitat World Health Organization: Definition and diagnosis of diabetes mellitus and intermediate hyperglycaemia: a report of a WHO/IDF consultation. 2006, Geneva World Health Organization: Definition and diagnosis of diabetes mellitus and intermediate hyperglycaemia: a report of a WHO/IDF consultation. 2006, Geneva
12.
Zurück zum Zitat Wolever TMS, Mehling C: Long-term effect of varying the source or amount of dietary carbohydrate on postprandial glucose, insulin, triacylglycerol, and free fatty acid concentrations in subjects with impaired glucose tolerance. Am J Clin Nutr. 2003, 77: 612-621. Wolever TMS, Mehling C: Long-term effect of varying the source or amount of dietary carbohydrate on postprandial glucose, insulin, triacylglycerol, and free fatty acid concentrations in subjects with impaired glucose tolerance. Am J Clin Nutr. 2003, 77: 612-621.
14.
Zurück zum Zitat Pruessner JC, Kirschbaum C, Meinlschmid G, Hellhammer DH: Two formulas for computation of area under the curve represent measures of total hormone concentration versus time-dependent change. Psychoneuroendocrinology. 2003, 28: 916-931. 10.1016/S0306-4530(02)00108-7.CrossRef Pruessner JC, Kirschbaum C, Meinlschmid G, Hellhammer DH: Two formulas for computation of area under the curve represent measures of total hormone concentration versus time-dependent change. Psychoneuroendocrinology. 2003, 28: 916-931. 10.1016/S0306-4530(02)00108-7.CrossRef
15.
Zurück zum Zitat Muniyappa R, Lee S, Chen H, Quon MJ: Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usages. Am J Physiol Endocrinol Metab. 2008, 294: E15-E26. 10.1152/ajpendo.00645.2007.CrossRef Muniyappa R, Lee S, Chen H, Quon MJ: Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usages. Am J Physiol Endocrinol Metab. 2008, 294: E15-E26. 10.1152/ajpendo.00645.2007.CrossRef
16.
Zurück zum Zitat Cassady BA, Hollis JH, Fulford AD, Considine RV, Mattes RD: Mastication of almonds: effects of lipid bioavailability, appetite, and hormone response. Am J Clin Nutr. 2009, 89: 794-800. 10.3945/ajcn.2008.26669.CrossRef Cassady BA, Hollis JH, Fulford AD, Considine RV, Mattes RD: Mastication of almonds: effects of lipid bioavailability, appetite, and hormone response. Am J Clin Nutr. 2009, 89: 794-800. 10.3945/ajcn.2008.26669.CrossRef
17.
Zurück zum Zitat Hunt JN, Stubbs DF: The volume and energy content of meals as determinants of gastric emptying. J Physiol. 1975, 245: 209-225.CrossRef Hunt JN, Stubbs DF: The volume and energy content of meals as determinants of gastric emptying. J Physiol. 1975, 245: 209-225.CrossRef
18.
Zurück zum Zitat Thompson LU, Yoon JH, Jenkins DJA, Wolever TMS, Jenkins AL: Relationship between polyphenol intake and blood glucose response of normal and diabetic individuals. Am J Clin Nutr. 1984, 39: 745-751. Thompson LU, Yoon JH, Jenkins DJA, Wolever TMS, Jenkins AL: Relationship between polyphenol intake and blood glucose response of normal and diabetic individuals. Am J Clin Nutr. 1984, 39: 745-751.
19.
Zurück zum Zitat Wolever TM, Bentum-Williams A, Jenkins DJ: Physiological modulation of plasma free fatty acid concentrations by diet. Metabolic implications in nondiabetic subjects. Diabetes Care. 1995, 18: 962-970. 10.2337/diacare.18.7.962.CrossRef Wolever TM, Bentum-Williams A, Jenkins DJ: Physiological modulation of plasma free fatty acid concentrations by diet. Metabolic implications in nondiabetic subjects. Diabetes Care. 1995, 18: 962-970. 10.2337/diacare.18.7.962.CrossRef
20.
Zurück zum Zitat Jenkins DJ, Wolever TM, Taylor RH: Slow release dietary carbohydrate improves second meal tolerance. Am J Clin Nutr. 1982, 35: 1339-1346. Jenkins DJ, Wolever TM, Taylor RH: Slow release dietary carbohydrate improves second meal tolerance. Am J Clin Nutr. 1982, 35: 1339-1346.
21.
Zurück zum Zitat Jackson KG, Wolstencroft EJ, Bateman PA, Yaqoob P, Williams CM: Acute effects of meal fatty acids on postprandial NEFA, glucose and apo E response: implications for insulin sensitivity and lipoprotein regulation?. Brit J Nutr. 2005, 93: 693-700. 10.1079/BJN20051410.CrossRef Jackson KG, Wolstencroft EJ, Bateman PA, Yaqoob P, Williams CM: Acute effects of meal fatty acids on postprandial NEFA, glucose and apo E response: implications for insulin sensitivity and lipoprotein regulation?. Brit J Nutr. 2005, 93: 693-700. 10.1079/BJN20051410.CrossRef
22.
Zurück zum Zitat Wachters-Hagedoorn RE, Priebe MG, Heimweg JAJ: The rate of intestinal glucose absorption is correlated with plasma glucose-dependent insulinotropic polypeptide concentrations in healthy men. J Nutr. 2006, 136: 1511-1516. Wachters-Hagedoorn RE, Priebe MG, Heimweg JAJ: The rate of intestinal glucose absorption is correlated with plasma glucose-dependent insulinotropic polypeptide concentrations in healthy men. J Nutr. 2006, 136: 1511-1516.
23.
Zurück zum Zitat Verdich C, Flint A, Gutzwiller E, Näslund E: A meta-analysis of the effect of glucagon-like peptide-1 (7-36) amide on ad libitum energy intake in humans. J Clin Endocrinol Metab. 2001, 86: 4382-4389. 10.1210/jc.86.9.4382. Verdich C, Flint A, Gutzwiller E, Näslund E: A meta-analysis of the effect of glucagon-like peptide-1 (7-36) amide on ad libitum energy intake in humans. J Clin Endocrinol Metab. 2001, 86: 4382-4389. 10.1210/jc.86.9.4382.
24.
Zurück zum Zitat Collier GR, Wolever TMS, Jenkins DJA: Concurrent ingestion of fat and reduction in starch content impairs carbohydrate tolerance to subsequent meals. Am J Clin Nutr. 1987, 45: 963-969. Collier GR, Wolever TMS, Jenkins DJA: Concurrent ingestion of fat and reduction in starch content impairs carbohydrate tolerance to subsequent meals. Am J Clin Nutr. 1987, 45: 963-969.
25.
Zurück zum Zitat Mandalari G, Faulks RM, Rich GT, Lo Turco V, Picout DR, Lo Curto RB, Bisignano G, Dugo P, Dugo G, Waldron KW, Ellis PR, Wickham MSJ: Release of protein, lipid, and vitamin E from almond seeds during digestion. J Ag Food Chem. 2008, 56: 3409-3416. 10.1021/jf073393v.CrossRef Mandalari G, Faulks RM, Rich GT, Lo Turco V, Picout DR, Lo Curto RB, Bisignano G, Dugo P, Dugo G, Waldron KW, Ellis PR, Wickham MSJ: Release of protein, lipid, and vitamin E from almond seeds during digestion. J Ag Food Chem. 2008, 56: 3409-3416. 10.1021/jf073393v.CrossRef
26.
Zurück zum Zitat Sakata T, Yoshimatsu H, Kurokawa M: Hypothalamic neuronal histamine: implications of its homeostatic control of energy metabolism. Nutr. 1997, 13: 403-411. 10.1016/S0899-9007(97)91277-6.CrossRef Sakata T, Yoshimatsu H, Kurokawa M: Hypothalamic neuronal histamine: implications of its homeostatic control of energy metabolism. Nutr. 1997, 13: 403-411. 10.1016/S0899-9007(97)91277-6.CrossRef
27.
Zurück zum Zitat Wolever TMS, Jenkins DJA, Ocana AM, Rao VA, Collier GR: Second-meal effect: low-glycemic index foods eaten at dinner improve subsequent breakfast glycemic response. Am J Clin Nutr. 1988, 48: 1041-1047. Wolever TMS, Jenkins DJA, Ocana AM, Rao VA, Collier GR: Second-meal effect: low-glycemic index foods eaten at dinner improve subsequent breakfast glycemic response. Am J Clin Nutr. 1988, 48: 1041-1047.
28.
Zurück zum Zitat Wascher TC, Schmoelzer I, Wiegratz A: Reduction of postchallenge hyperglycaemia prevents acute endothelial dysfunction in subjects with impaired glucose tolerance. Eur J Clin Invest. 2005, 35: 551-557. 10.1111/j.1365-2362.2005.01550.x.CrossRef Wascher TC, Schmoelzer I, Wiegratz A: Reduction of postchallenge hyperglycaemia prevents acute endothelial dysfunction in subjects with impaired glucose tolerance. Eur J Clin Invest. 2005, 35: 551-557. 10.1111/j.1365-2362.2005.01550.x.CrossRef
Metadaten
Titel
Acute and second-meal effects of almond form in impaired glucose tolerant adults: a randomized crossover trial
verfasst von
Alisa M Mori
Robert V Considine
Richard D Mattes
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Nutrition & Metabolism / Ausgabe 1/2011
Elektronische ISSN: 1743-7075
DOI
https://doi.org/10.1186/1743-7075-8-6

Weitere Artikel der Ausgabe 1/2011

Nutrition & Metabolism 1/2011 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Frühzeitige HbA1c-Kontrolle macht sich lebenslang bemerkbar

22.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes von Anfang an intensiv BZ-senkend zu behandeln, wirkt sich positiv auf Komplikationen und Mortalität aus – und das offenbar lebenslang, wie eine weitere Nachfolgeuntersuchung der UKPD-Studie nahelegt.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.