Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Liselotte van Bloemendaal is active.

Publication


Featured researches published by Liselotte van Bloemendaal.


Diabetes Care | 2015

Liraglutide Reduces CNS Activation in Response to Visual Food Cues Only After Short-term Treatment in Patients With Type 2 Diabetes

Jennifer S. ten Kulve; Dick J. Veltman; Liselotte van Bloemendaal; Frederik Barkhof; Madeleine L. Drent; Michaela Diamant; Richard G. IJzerman

OBJECTIVE Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are associated with reduced appetite and body weight. We investigated whether these effects could be mediated by the central nervous system (CNS). RESEARCH DESIGN AND METHODS We performed a randomized crossover study in obese patients with type 2 diabetes (n = 20, mean age 59.3 ± 4.1 years, mean BMI 32 ± 4.7 kg/m2), consisting of two periods of 12-week treatment with either liraglutide 1.8 mg or insulin glargine. Using functional MRI, we determined the effects of treatment on CNS responses to viewing food pictures in the fasted condition and 30 min after meal intake. RESULTS After 12 weeks, the decrease in HbA1c was larger with liraglutide versus insulin glargine (Δ−0.7% vs. −0.2%, P < 0.001). Body weight decreased during liraglutide versus insulin glargine (Δ−3.3 kg vs. 0.8 kg, P < 0.001). After 10 days, patients treated with liraglutide, compared with insulin glargine, showed decreased responses to food pictures in insula and putamen (P ≤ 0.02). In addition, liraglutide enhanced the satiating effect of meal intake on responses in putamen and amygdala (P ≤ 0.05). Differences between liraglutide and insulin glargine were not observed after 12 weeks. CONCLUSIONS Compared with insulin, liraglutide decreased CNS activation significantly only after short-term treatment, suggesting that these effects of GLP-1RA on the CNS may contribute to the induction of weight loss, but not necessarily to its maintenance, in view of the absence of an effect of liraglutide on CNS activation in response to food pictures after longer-term treatment.


Metabolic Brain Disease | 2016

Alterations in white matter volume and integrity in obesity and type 2 diabetes.

Liselotte van Bloemendaal; Richard G. IJzerman; Jennifer S. ten Kulve; Frederik Barkhof; Michaela Diamant; Dick J. Veltman; Eelco van Duinkerken

Type 2 diabetes mellitus (T2DM) is characterized by obesity, hyperglycemia and insulin resistance. Both T2DM and obesity are associated with cerebral complications, including an increased risk of cognitive impairment and dementia, however the underlying mechanisms are largely unknown. In the current study, we aimed to determine the relative contributions of obesity and the presence of T2DM to altered white matter structure. We used diffusion tensor imaging (DTI) and voxel-based morphometry (VBM) to measure white matter integrity and volume in obese T2DM patients without micro- or macrovascular complications, age- gender- and BMI-matched normoglycemic obese subjects and age- and gender-matched normoglycemic lean subjects. We found that obese T2DM patients compared with lean subjects had lower axial diffusivity (in the right corticospinal tract, right inferior fronto-occipital tract, right superior longitudinal fasciculus and right forceps major) and reduced white matter volume (in the right inferior parietal lobe and the left external capsule region). In normoglycemic obese compared with lean subjects axial diffusivity as well as white matter volume tended to be reduced, whereas there were no significant differences between normoglycemic obese subjects and T2DM patients. Decreased white matter integrity and volume were univariately related to higher age, being male, higher BMI, HbA1C and fasting glucose and insulin levels. However, multivariate analyses demonstrated that only BMI was independently related to white matter integrity, and age, gender and BMI to white matter volume loss. Our data indicate that obese T2DM patients have reduced white matter integrity and volume, but that this is largely explained by BMI, rather than T2DM per se.


Journal of Endocrinology | 2016

Endogenous GLP1 and GLP1 analogue alter CNS responses to palatable food consumption

Jennifer S. Ten Kulve; Dick J. Veltman; Liselotte van Bloemendaal; Paul Groot; Henricus G. Ruhé; Frederik Barkhof; Michaela Diamant; Richard G. IJzerman

Glucagon-like peptide-1 (GLP1) affects appetite, supposedly mediated via the central nervous system (CNS). In this study, we investigate whether modulation of CNS responses to palatable food consumption may be a mechanism by which GLP1 contributes to the central regulation of feeding. Using functional MRI, we determined the effects of endogenous GLP1 and treatment with the GLP1 analogue liraglutide on CNS activation to chocolate milk receipt. Study 1 included 20 healthy lean individuals and 20 obese patients with type 2 diabetes (T2DM). Scans were performed on two occasions: during infusion of the GLP1 receptor antagonist exendin 9-39 (blocking actions of endogenous GLP1) and during placebo infusion. Study 2 was a randomised, cross-over intervention study carried out in 20 T2DM patients, comparing treatment with liraglutide to insulin, after 10 days and 12 weeks. Compared with lean individuals, T2DM patients showed reduced activation to chocolate milk in right insula (P = 0.04). In lean individuals, blockade of endogenous GLP1 effects inhibited activation in bilateral insula (P ≤ 0.03). Treatment in T2DM with liraglutide, vs insulin, increased activation to chocolate milk in right insula and caudate nucleus after 10 days (P ≤ 0.03); however, these effects ceased to be significant after 12 weeks. Our findings in healthy lean individuals indicate that endogenous GLP1 is involved in the central regulation of feeding by affecting central responsiveness to palatable food consumption. In obese T2DM, treatment with liraglutide may improve the observed deficit in responsiveness to palatable food, which may contribute to the induction of weight loss observed during treatment. However, no long-term effects of liraglutide were observed.


The Journal of Clinical Endocrinology and Metabolism | 2016

Decreased Hypothalamic Glucagon-Like Peptide-1 Receptor Expression in Type 2 Diabetes Patients

Jennifer S. ten Kulve; Liselotte van Bloemendaal; Rawien Balesar; Richard G. IJzerman; Dick F. Swaab; Michaela Diamant; Susanne E. la Fleur; Anneke Alkemade

CONTEXT Glucagon-like peptide-1 (GLP-1) and GLP-1 receptor agonist treatment in type 2 diabetes (T2DM) reduce blood glucose and food intake. It has been suggested that these effects are partly mediated through central GLP-1 receptors (GLP-1Rs). The rodent and nonhuman primate hypothalamus show clear GLP-1R expression. However, a detailed description of GLP-1R expression in the human hypothalamus is lacking, and it is unknown whether this expression is altered in T2DM patients. OBJECTIVE The objective of the study was to investigate the GLP-1R distribution in the human postmortem hypothalamus and to determine whether hypothalamic GLP-1R expression is altered in T2DM patients. DESIGN We investigated the distribution of GLP-1R expression throughout the human hypothalamus by means of in situ hybridization. We also performed quantifications of GLP-1R mRNA expression in two hypothalamic nuclei (ie, the paraventricular nucleus [PVN] and infundibular nucleus [IFN]), comparing patients with T2DM and control subjects. RESULTS We found that GLP-1R mRNA was expressed in a number of hypothalamic nuclei including the PVN and the IFN, both involved in the regulation of energy metabolism. We observed sporadic colocalization of the GLP-1R in the IFN with the orgexigenic neuropeptide Y, agouti-related peptide, or proopiomelanocortin transcripts. Comparison of GLP-1R mRNA in the PVN and IFN between T2DM patients and control subjects revealed a decreased expression in T2DM patients. CONCLUSIONS Our studies show that GLP-1R is widely expressed throughout the human hypothalamus. The decreased expression of GLP-1R in the PVN and IFN of T2DM patients may be related to the dysregulation of feeding behavior and glucose homeostasis in T2DM.


Diabetes Care | 2017

Elevated Postoperative Endogenous GLP-1 Levels Mediate Effects of Roux-en-Y Gastric Bypass on Neural Responsivity to Food Cues

Jennifer S. ten Kulve; Dick J. Veltman; Victor E. A. Gerdes; Liselotte van Bloemendaal; Frederik Barkhof; Carolyn F. Deacon; Jens J. Holst; Madeleine L. Drent; Michaela Diamant; Richard G. IJzerman

OBJECTIVE It has been suggested that weight reduction and improvements in satiety after Roux-en-Y gastric bypass (RYGB) are partly mediated via postoperative neuroendocrine changes. Glucagon-like peptide-1 (GLP-1) is a gut hormone secreted after food ingestion and is associated with appetite and weight reduction, mediated via effects on the central nervous system (CNS). Secretion of GLP-1 is greatly enhanced after RYGB. We hypothesized that postoperative elevated GLP-1 levels contribute to the improved satiety regulation after RYGB via effects on the CNS. RESEARCH DESIGN AND METHODS Effects of the GLP-1 receptor antagonist exendin 9-39 (Ex9-39) and placebo were assessed in 10 women before and after RYGB. We used functional MRI to investigate CNS activation in response to visual food cues (pictures) and gustatory food cues (consumption of chocolate milk), comparing results with Ex9-39 versus placebo before and after RYGB. RESULTS After RYGB, CNS activation was reduced in the rolandic operculum and caudate nucleus in response to viewing food pictures (P = 0.03) and in the insula in response to consumption of palatable food (P = 0.003). GLP-1 levels were significantly elevated postoperatively (P < 0.001). After RYGB, GLP-1 receptor blockade resulted in a larger increase in activation in the caudate nucleus in response to food pictures (P = 0.02) and in the insula in response to palatable food consumption (P = 0.002). CONCLUSIONS We conclude that the effects of RYGB on CNS activation in response to visual and gustatory food cues may be mediated by central effects of GLP-1. Our findings provide further insights into the mechanisms underlying the weight-lowering effects of RYGB.


Diabetes Care | 2018

SGLT2 Inhibitors in Combination Therapy: From Mechanisms to Clinical Considerations in Type 2 Diabetes Management

Michaël J.B. van Baar; Charlotte C. van Ruiten; Marcel H.A. Muskiet; Liselotte van Bloemendaal; Richard G. IJzerman; Daniël H. van Raalte

The progressive nature of type 2 diabetes (T2D) requires practitioners to periodically evaluate patients and intensify glucose-lowering treatment once glycemic targets are not attained. With guidelines moving away from a one-size-fits-all approach toward setting patient-centered goals and allowing flexibility in choosing a second-/third-line drug from the growing number of U.S. Food and Drug Administration–approved glucose-lowering agents, keen personalized management in T2D has become a challenge for health care providers in daily practice. Among the newer generation of glucose-lowering drug classes, sodium–glucose cotransporter 2 inhibitors (SGLT2is), which enhance urinary glucose excretion to lower hyperglycemia, have made an imposing entrance to the T2D treatment armamentarium. Given their unique insulin-independent mode of action and their favorable efficacy–to–adverse event profile and given their marked benefits on cardiovascular-renal outcome in moderate-to-high risk T2D patients, which led to updates of guidelines and product monographs, the role of this drug class in multidrug regimes is promising. However, despite many speculations based on pharmacokinetic and pharmacodynamic properties, physiological reasoning, and potential synergism, the effects of these agents in terms of glycemic and pleiotropic efficacy when combined with other glucose-lowering drug classes are largely understudied. In this perspective, we review the currently emerging evidence, discuss prevailing hypotheses, and elaborate on necessary future studies to clarify the potential risks and benefits of using an SGLT2i in dual combination with metformin and triple combination with a glucagon-like peptide 1 receptor agonist, dipeptidyl peptidase 4 inhibitor, or other glucose-lowering agent that is recommended by the American Diabetes Association and European Association for the Study of Diabetes (i.e., a sulfonylurea, thiazolidinedione, or insulin) to treat patients with T2D.


Metabolic Brain Disease | 2018

Cortical and subcortical gray matter structural alterations in normoglycemic obese and type 2 diabetes patients: relationship with adiposity, glucose, and insulin

Gabriel Bernardes; Richard G. IJzerman; Jennifer S. ten Kulve; Frederik Barkhof; Michaela Diamant; Dick J. Veltman; J. Landeira-Fernandez; Liselotte van Bloemendaal; Eelco van Duinkerken

Type 2 diabetes (T2DM) is associated with structural cortical and subcortical alterations, although it is insufficiently clear if these alterations are driven by obesity or by diabetes and its associated complications. We used FreeSurfer5.3 and FSL-FIRST to determine cortical thickness, volume and surface area, and subcortical gray matter volume in a group of 16 normoglycemic obese subjects and 28 obese T2DM patients without clinically manifest micro- and marcoangiopathy, and compared them to 31 lean normoglycemic controls. Forward regression analysis was used to determine demographic and clinical correlates of altered (sub)cortical structure. Exploratively, vertex-wise correlations between cortical structure and fasting glucose and insulin were calculated. Compared with controls, obese T2DM patients showed lower right insula thickness and lower left lateral occipital surface area (PFWE < 0.05). Normoglycemic obese versus controls had lower thickness (PFWE < 0.05) in the right insula and inferior frontal gyrus, and higher amygdala and thalamus volume. Thalamus volume and left paracentral surface area were also higher in this group compared with obese T2DM patients. Age, sex, BMI, fasting glucose, and cholesterol were related to these (sub)cortical alterations in the whole group (all P < 0.05). Insulin were related to temporal and frontal structural deficits (all PFWE < 0.05). Parietal/occipital structural deficits may constitute early T2DM-related cerebral alterations, whereas in normoglycemic obese subjects, regions involved in emotion, appetite, satiety regulation, and inhibition were affected. Central adiposity and elevated fasting glucose may constitute risk factors.


Nederlands Tijdschrift voor Diabetologie | 2015

14. Liraglutide Decreases Food Related CNS Activation after Short-Term, but Not after Longer-Term Treatment in Patients with Diabetes (159-OR)

Jennifer S. Ten Kulve; Dick J. Veltman; Liselotte van Bloemendaal; Frederik Barkhof; Madeleine L. Drent; Michaela Diamant; Richard G. IJzerman

SamenvattingThe central nervous system (CNS) is a major player in the regulation of feeding. Gut-derived hormones, such as glucagon- like peptide-1 (GLP-1), may relay information about the nutritional status to the CNS. Treatment with GLP-1 agonists, such as liraglutide, is associated with reduced appetite and weight loss. We hypothesized that these effects are mediated by effects on the CNS. We performed a randomized cross-over study in patients with type 2 diabetes (n = 20, mean ± SD, age 59.3 ± 4.1 yr, BMI 32 ± 4.7 kg/m2). Each individual underwent 2 periods of 12 week treatment with either liraglutide or insulin glargine with a 12 week wash-out period. Using fMRI, we determined the effects of treatment on CNS activation to food vs. nonfood pictures in areas regulating reward when fasted and 30 min. after a standardized meal. FMRI scans were performed at baseline, after 10 days and 12 weeks of treatment. After 12 weeks, the decrease in HbA1c was larger with liraglutide vs. insulin (Δ -0.7% vs. -0.2%, p difference < 0.001). Body weight decreased during liraglutide vs insulin (Δ -3.3 kg vs. +0.8 kg, p difference < 0.001). After 10 days, before weight changes had occurred, patients treated with liraglutide, compared to insulin, showed decreased activation to food pictures in left putamen in the postprandial condition. After 10 days, in comparison with insulin, liraglutide enhanced the reducing effect of the meal intake on CNS activation to food pictures in left putamen and amygdala. These differences between liraglutide and insulin were not observed after 12 weeks. Compared to insulin, liraglutide decreased CNS activations in response to food pictures in reward areas after short term, but not after longer term treatment. Our findings suggest that the GLP-1 mediated altered CNS activation may contribute to the induction of weight loss. However the absence of effects after longer term treatment might explain why weight loss does not proceed after the initial treatment period with liraglutide.


Nederlands Tijdschrift voor Diabetologie | 2015

24. Brain Reward-System Activation in Response to Anticipation and Consumption of Palatable Food Is Altered by GLP-1 Receptor Activation in Humans (384-OR)

Liselotte van Bloemendaal; Dick J. Veltman; Jennifer S. Ten Kulve; Paul Groot; Henricus G. Ruhé; Frederik Barkhof; John H. Sloan; Michaela Diamant; Richard G. IJzerman

SamenvattingIt has been suggested that obese individuals have increased brain reward system activation while anticipating food intake, which may lead to cravings for food, and decreased reward system activation during actual food consumption, which may induce overeating. Gut-derived hormones, such as glucagon-like peptide-1 (GLP-1), are likely involved in the regulation of food intake. GLP-1 receptor agonists improve glycemic control and reduce food intake and body weight. We hypothesized that food intake reduction following GLP-1 receptor activation is mediated through brain areas regulating anticipatory and consummatory food reward. Using functional MRI, we determined the effects of GLP-1 receptor activation on brain responses to anticipation and receipt of chocolate milk vs. tasteless solution. Obese T2DM patients, normoglycemic obese and lean subjects (n = 48) underwent three functional MRI sessions at separate visits with intravenous infusion of A) the GLP-1 receptor agonist exenatide, B) exenatide with prior GLP-1 receptor blockade by exendin 9-39 or C) placebo; during somatostatin pituitary-pancreatic clamps. We found that BMI negatively correlated with brain responses to receipt of chocolate milk and positively correlated with anticipation of receipt of chocolate milk in brain areas regulating reward, appetite and motivation. Exenatide vs. placebo increased brain responses to receipt of chocolate milk and decreased anticipation of receipt of chocolate milk, paralleled by reductions in food intake. Exendin 9-39 largely prevented these effects. GLP-1 receptor activation decreased anticipatory food reward, which may reduce cravings for food, and increased consummatory food reward, which may prevent overeating. Our findings provide novel insights into the mechanisms by which GLP-1 regulates food intake and how GLP-1 receptor agonists induce weight loss.


Nederlands Tijdschrift voor Diabetologie | 2013

PS18 - 1. Exenatide blunts hyperactivation in CNS reward and satiety circuits elicited by viewing food cues in obese individuals

Liselotte van Bloemendaal; Richard G. IJzerman; Jennifer S. Ten Kulve; Frederik Barkhof; Dick J. Veltman; Michaela Diamant

The GLP-1 receptor agonist (GLP-1RA) exenatide improves glycaemic control and promotes satiety, leading to reductions in food intake and body weight. GLP-1RA actions on the brain may partly mediate satiety and weight effects. We hypothesised that exenatide reduces food intake by affecting CNS reward and satiety circuits.

Collaboration


Dive into the Liselotte van Bloemendaal's collaboration.

Top Co-Authors

Avatar

Richard G. IJzerman

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dick J. Veltman

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michaela Diamant

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Madeleine L. Drent

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eelco van Duinkerken

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge