Network


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

Hotspot


Dive into the research topics where Willem Staels is active.

Publication


Featured researches published by Willem Staels.


Diabetes | 2015

Estrogen Receptor α Regulates β-Cell Formation During Pancreas Development and Following Injury

Yixing Yuchi; Ying Cai; Bart Legein; Sofie De Groef; Gunter Leuckx; Violette Coppens; Eva Van Overmeire; Willem Staels; Nico De Leu; Geert A. Martens; Jo A. Van Ginderachter; Harry Heimberg; Mark Van de Casteele

Identifying pathways for β-cell generation is essential for cell therapy in diabetes. We investigated the potential of 17β-estradiol (E2) and estrogen receptor (ER) signaling for stimulating β-cell generation during embryonic development and in the severely injured adult pancreas. E2 concentration, ER activity, and number of ERα transcripts were enhanced in the pancreas injured by partial duct ligation (PDL) along with nuclear localization of ERα in β-cells. PDL-induced proliferation of β-cells depended on aromatase activity. The activation of Neurogenin3 (Ngn3) gene expression and β-cell growth in PDL pancreas were impaired when ERα was turned off chemically or genetically (ERα−/−), whereas in situ delivery of E2 promoted β-cell formation. In the embryonic pancreas, β-cell replication, number of Ngn3+ progenitor cells, and expression of key transcription factors of the endocrine lineage were decreased by ERα inactivation. The current study reveals that E2 and ERα signaling can drive β-cell replication and formation in mouse pancreas.


Journal of Affective Disorders | 2016

The effect of mood-stabilizing drugs on cytokine levels in bipolar disorder: A systematic review.

Seline van den Ameele; Linda van Diermen; Willem Staels; Violette Coppens; Glenn Dumont; Bernard Sabbe; Manuel Morrens

OBJECTIVES Cytokine level alterations suggest a role for the immune system in the pathophysiology of bipolar disorder (BD). Pharmacotherapy is an important confounding factor in clinical research on cytokine levels. In this systematic review we collate the evidence on blood cytokine levels in medication-free BD and the effects of single mood-stabilizing drugs on these levels. METHODS A systematic review was conducted according to the PRISMA statement. We searched the Pubmed and Embase databases for clinical studies reporting either on cytokine levels in medication-free BD or on the effects of single mood-stabilizing drugs on cytokine levels in BD. RESULTS Of the 564 articles screened, 17 were included. Fourteen articles report on medication-free patients with BD and indicate state-related cytokine alterations. Six articles discuss the effect of lithium. Whereas no data on short-term effects of lithium were found, ≥2 months lithium use in euthymic populations is associated with normal cytokine levels. Two studies report no effect of valproate and no studies were found on carbamazepine, lamotrigine or antipsychotics. LIMITATIONS The available studies are characterized by a broad methodological heterogeneity and limited replication between studies. CONCLUSIONS This systematic review suggests the presence of state-related cytokine level alterations in medication-free BD with most evidence pointing to a proinflammatory cytokine response in mania. Euthymia and long-term lithium use are associated with normal cytokine levels. To improve our understanding of the impact of mood-stabilizing drugs on cytokine levels, longitudinal studies with medication-free baseline, randomized controlled single-drug treatment protocols and close mood state monitoring are needed.


Stem Cells Translational Medicine | 2015

Concise Review: Macrophages: Versatile Gatekeepers During Pancreatic β-Cell Development, Injury, and Regeneration

Naomi Van Gassen; Willem Staels; Eva Van Overmeire; Sofie De Groef; Mozhdeh Sojoodi; Yves Heremans; Gunter Leuckx; Mark Van de Casteele; Jo A. Van Ginderachter; Harry Heimberg; Nico De Leu

Macrophages are classically considered detrimental for pancreatic β‐cell survival and function, thereby contributing to β‐cell failure in both type 1 (T1D) and 2 (T2D) diabetes mellitus. In addition, adipose tissue macrophages negatively influence peripheral insulin signaling and promote obesity‐induced insulin resistance in T2D. In contrast, recent data unexpectedly uncovered that macrophages are not only able to protect β cells during pancreatitis but also to orchestrate β‐cell proliferation and regeneration after β‐cell injury. Moreover, by altering their activation state, macrophages are able to improve insulin resistance in murine models of T2D. This review will elaborate on current insights in macrophage heterogeneity and on the evolving role of pancreas macrophages during organogenesis, tissue injury, and repair. Additional identification of macrophage subtypes and of their secreted factors might ultimately translate into novel therapeutic strategies for both T1D and T2D.


Diabetes, Obesity and Metabolism | 2016

Accessory cells for β-cell transplantation.

Willem Staels; S. De Groef; Yves Heremans; Violette Coppens; N. Van Gassen; Gunter Leuckx; M Van de Casteele; I. Van Riet; Aernout Luttun; Harry Heimberg; N. De Leu

Despite recent advances, insulin therapy remains a treatment, not a cure, for diabetes mellitus with persistent risk of glycaemic alterations and life‐threatening complications. Restoration of the endogenous β‐cell mass through regeneration or transplantation offers an attractive alternative. Unfortunately, signals that drive β‐cell regeneration remain enigmatic and β‐cell replacement therapy still faces major hurdles that prevent its widespread application. Co‐transplantation of accessory non‐islet cells with islet cells has been shown to improve the outcome of experimental islet transplantation. This review will highlight current travails in β‐cell therapy and focuses on the potential benefits of accessory cells for islet transplantation in diabetes.


Diabetes | 2018

Inhibition of Cdk5 promotes β-cell differentiation from ductal progenitors

Ka-Cheuk Liu; Gunter Leuckx; Daisuke Sakano; Philip A. Seymour; Charlotte L. Mattsson; Linn Rautio; Willem Staels; Yannick Verdonck; Palle Serup; Shoen Kume; Harry Heimberg; Olov Andersson

Inhibition of notch signaling is known to induce differentiation of endocrine cells in zebrafish and mouse. After performing an unbiased in vivo screen of ∼2,200 small molecules in zebrafish, we identified an inhibitor of Cdk5 (roscovitine), which potentiated the formation of β-cells along the intrapancreatic duct during concurrent inhibition of notch signaling. We confirmed and characterized the effect with a more selective Cdk5 inhibitor, (R)-DRF053, which specifically increased the number of duct-derived β-cells without affecting their proliferation. By duct-specific overexpression of the endogenous Cdk5 inhibitors Cdk5rap1 or Cdkal1 (which previously have been linked to diabetes in genome-wide association studies), as well as deleting cdk5, we validated the role of chemical Cdk5 inhibition in β-cell differentiation by genetic means. Moreover, the cdk5 mutant zebrafish displayed an increased number of β-cells independently of inhibition of notch signaling, in both the basal state and during β-cell regeneration. Importantly, the effect of Cdk5 inhibition to promote β-cell formation was conserved in mouse embryonic pancreatic explants, adult mice with pancreatic ductal ligation injury, and human induced pluripotent stem (iPS) cells. Thus, we have revealed a previously unknown role of Cdk5 as an endogenous suppressor of β-cell differentiation and thereby further highlighted its importance in diabetes.


Journal of Visualized Experiments | 2015

Surgical Injury to the Mouse Pancreas through Ligation of the Pancreatic Duct as a Model for Endocrine and Exocrine Reprogramming and Proliferation.

Sofie De Groef; Gunter Leuckx; Naomi Van Gassen; Willem Staels; Ying Cai; Yixing Yuchi; Violette Coppens; Nico De Leu; Yves Heremans; Luc Baeyens; Mark Van de Casteele; Harry Heimberg

Expansion of pancreatic beta cells in vivo or ex vivo, or generation of beta cells by differentiation from an embryonic or adult stem cell, can provide new expandable sources of beta cells to alleviate the donor scarcity in human islet transplantation as therapy for diabetes. Although recent advances have been made towards this aim, mechanisms that regulate beta cell expansion and differentiation from a stem/progenitor cell remain to be characterized. Here, we describe a protocol for an injury model in the adult mouse pancreas that can function as a tool to study mechanisms of tissue remodeling and beta cell proliferation and differentiation. Partial duct ligation (PDL) is an experimentally induced injury of the rodent pancreas involving surgical ligation of the main pancreatic duct resulting in an obstruction of drainage of exocrine products out of the tail region of the pancreas. The inflicted damage induces acinar atrophy, immune cell infiltration and severe tissue remodeling. We have previously reported the activation of Neurogenin (Ngn) 3 expressing endogenous progenitor-like cells and an increase in beta cell proliferation after PDL. Therefore, PDL provides a basis to study signals involved in beta cell dynamics and the properties of an endocrine progenitor in adult pancreas. Since, it still remains largely unclear, which factors and pathways contribute to beta cell neogenesis and proliferation in PDL, a standardized protocol for PDL will allow for comparison across laboratories.


Cell Death and Disease | 2016

STAT3 modulates β-cell cycling in injured mouse pancreas and protects against DNA damage

S. De Groef; D Renmans; Ying Cai; Gunter Leuckx; S Roels; Willem Staels; Gérard Gradwohl; Luc Baeyens; Yves Heremans; Geert A. Martens; N. De Leu; Mozhdeh Sojoodi; M Van de Casteele; Harry Heimberg

Partial pancreatic duct ligation (PDL) of mouse pancreas induces a doubling of the β-cell mass mainly through proliferation of pre-existing and newly formed β-cells. The molecular mechanism governing this process is still largely unknown. Given the inflammatory nature of PDL and inflammation-induced signaling via the signal transducer and activator of transcription 3 (STAT3), the activation and the role of STAT3 in PDL-induced β-cell proliferation were investigated. Duct ligation stimulates the expression of several cytokines that can act as ligands inducing STAT3 signaling and phosphorylation in β-cells. β-Cell cycling increased by conditional β-cell-specific Stat3 knockout and decreased by STAT3 activation through administration of interleukin-6. In addition, the level of DNA damage in β-cells of PDL pancreas increased after deletion of Stat3. These data indicate a role for STAT3 in maintaining a steady state in the β-cell, by modulating its cell cycle and protection from DNA damage.


Best Practice & Research Clinical Endocrinology & Metabolism | 2015

Reprogramming of human exocrine pancreas cells to beta cells

Willem Staels; Yves Heremans; Harry Heimberg

One of the key promises of regenerative medicine is providing a cure for diabetes. Cell-based therapies are proving their safety and efficiency, but donor beta cell shortages and immunological issues remain major hurdles. Reprogramming of human pancreatic exocrine cells towards beta cells would offer a major advantage by providing an abundant and autologous source of beta cells. Over the past decade our understanding of transdifferentiation processes greatly increased allowing us to design reprogramming protocols that fairly aim for clinical trials.


Physiological Reviews | 2018

(Re)generating Human Beta Cells: Status, Pitfalls, and Perspectives

Luc Baeyens; Marie Lemper; Willem Staels; Sofie De Groef; Nico De Leu; Yves Heremans; Michael S. German; Harry Heimberg

Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.


Diabetologia | 2018

Vegf-A mRNA transfection as a novel approach to improve mouse and human islet graft revascularisation

Willem Staels; Yannick Verdonck; Yves Heremans; Gunter Leuckx; Sofie De Groef; Carlo Heirman; Eelco J.P. de Koning; Conny Gysemans; Kris Thielemans; Luc Baeyens; Harry Heimberg; Nico De Leu

Aims/hypothesisThe initial avascular period following islet transplantation seriously compromises graft function and survival. Enhancing graft revascularisation to improve engraftment has been attempted through virus-based delivery of angiogenic triggers, but risks associated with viral vectors have hampered clinical translation. In vitro transcribed mRNA transfection circumvents these risks and may be used for improving islet engraftment.MethodsMouse and human pancreatic islet cells were transfected with mRNA encoding the angiogenic growth factor vascular endothelial growth factor A (VEGF-A) before transplantation under the kidney capsule in mice.ResultsAt day 7 post transplantation, revascularisation of grafts transfected with Vegf-A (also known as Vegfa) mRNA was significantly higher compared with non-transfected or Gfp mRNA-transfected controls in mouse islet grafts (2.11- and 1.87-fold, respectively) (vessel area/graft area, mean ± SEM: 0.118 ± 0.01 [n = 3] in Vegf-A mRNA transfected group (VEGF) vs 0.056 ± 0.01 [n = 3] in no RNA [p < 0.05] vs 0.063 ± 0.02 [n = 4] in Gfp mRNA transfected group (GFP) [p < 0.05]); EndoC-bH3 grafts (2.85- and 2.48-fold. respectively) (0.085 ± 0.02 [n = 4] in VEGF vs 0.030 ± 0.004 [n = 4] in no RNA [p < 0.05] vs 0.034 ± 0.01 [n = 5] in GFP [p < 0.05]); and human islet grafts (3.17- and 3.80-fold, respectively) (0.048 ± 0.013 [n = 3] in VEGF vs 0.015 ± 0.0051 [n = 4] in no RNA [p < 0.01] vs 0.013 ± 0.0046 [n = 4] in GFP [p < 0.01]). At day 30 post transplantation, human islet grafts maintained a vascularisation benefit (1.70- and 1.82-fold, respectively) (0.049 ± 0.0042 [n = 8] in VEGF vs 0.029 ± 0.0052 [n = 5] in no RNA [p < 0.05] vs 0.027 ± 0.0056 [n = 4] in GFP [p < 0.05]) and a higher beta cell volume (1.64- and 2.26-fold, respectively) (0.0292 ± 0.0032 μl [n = 7] in VEGF vs 0.0178 ± 0.0021 μl [n = 5] in no RNA [p < 0.01] vs 0.0129 ± 0.0012 μl [n = 4] in GFP [p < 0.001]).Conclusions/interpretationVegf-A mRNA transfection before transplantation provides a promising and safe strategy to improve engraftment of islets and other cell-based implants.

Collaboration


Dive into the Willem Staels's collaboration.

Top Co-Authors

Avatar

Harry Heimberg

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Gunter Leuckx

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Yves Heremans

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Nico De Leu

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Luc Baeyens

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Sofie De Groef

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mozhdeh Sojoodi

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar

Naomi Van Gassen

Vrije Universiteit Brussel

View shared research outputs
Researchain Logo
Decentralizing Knowledge