Network


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

Hotspot


Dive into the research topics where Emilie Castermans is active.

Publication


Featured researches published by Emilie Castermans.


Nature Methods | 2009

Parallel detection of antigen-specific T-cell responses by multidimensional encoding of MHC multimers.

Sine Reker Hadrup; Arnold H. Bakker; Chengyi J Shu; Rikke Andersen; Jerre van Veluw; Pleun Hombrink; Emilie Castermans; Per thor Straten; Christian U. Blank; John B. A. G. Haanen; Mirjam H.M. Heemskerk; Ton N. M. Schumacher

The use of fluorescently labeled major histocompatibility complex multimers has become an essential technique for analyzing disease- and therapy-induced T-cell immunity. Whereas classical major histocompatibility complex multimer analyses are well-suited for the detection of immune responses to a few epitopes, limitations on human-subject sample size preclude a comprehensive analysis of T-cell immunity. To address this issue, we developed a combinatorial encoding strategy that allows the parallel detection of a multitude of different T-cell populations in a single sample. Detection of T cells from peripheral blood by combinatorial encoding is as efficient as detection with conventionally labeled multimers but results in a substantially increased sensitivity and, most notably, allows comprehensive screens to be performed. We obtained proof of principle for the feasibility of large-scale screening of human material by analysis of human leukocyte antigen A3–restricted T-cell responses to known and potential melanoma-associated antigens in peripheral blood from individuals with melanoma.


Endocrine-related Cancer | 2015

X-linked acrogigantism syndrome: clinical profile and therapeutic responses.

Albert Beckers; Maya Lodish; Giampaolo Trivellin; Liliya Rostomyan; Misu Lee; Fabio R. Faucz; Bo Yuan; Catherine S. Choong; Jean-Hubert Caberg; Elisa Verrua; Luciana A. Naves; Tim Cheetham; Jacques Young; Philippe A. Lysy; Patrick Petrossians; Andrew Cotterill; Nalini S. Shah; Daniel Metzger; Emilie Castermans; Maria Rosaria Ambrosio; Chiara Villa; Natalia Strebkova; Nadia Mazerkina; Stephan Gaillard; Gustavo Barcelos Barra; Luis Augusto Casulari; Sebastian Neggers; Roberto Salvatori; Marie Lise Jaffrain-Rea; Margaret Zacharin

X-linked acrogigantism (X-LAG) is a new syndrome of pituitary gigantism, caused by microduplications on chromosome Xq26.3, encompassing the gene GPR101, which is highly upregulated in pituitary tumors. We conducted this study to explore the clinical, radiological, and hormonal phenotype and responses to therapy in patients with X-LAG syndrome. The study included 18 patients (13 sporadic) with X-LAG and microduplication of chromosome Xq26.3. All sporadic cases had unique duplications and the inheritance pattern in two families was dominant, with all Xq26.3 duplication carriers being affected. Patients began to grow rapidly as early as 2-3 months of age (median 12 months). At diagnosis (median delay 27 months), patients had a median height and weight standard deviation scores (SDS) of >+3.9 SDS. Apart from the increased overall body size, the children had acromegalic symptoms including acral enlargement and facial coarsening. More than a third of cases had increased appetite. Patients had marked hypersecretion of GH/IGF1 and usually prolactin, due to a pituitary macroadenoma or hyperplasia. Primary neurosurgical control was achieved with extensive anterior pituitary resection, but postoperative hypopituitarism was frequent. Control with somatostatin analogs was not readily achieved despite moderate to high levels of expression of somatostatin receptor subtype-2 in tumor tissue. Postoperative use of adjuvant pegvisomant resulted in control of IGF1 in all five cases where it was employed. X-LAG is a new infant-onset gigantism syndrome that has a severe clinical phenotype leading to challenging disease management.


Haematologica | 2011

Thymic recovery after allogeneic hematopoietic cell transplantation with non-myeloablative conditioning is limited to patients younger than 60 years of age

Emilie Castermans; Muriel Hannon; Jacques Dutrieux; Stéphanie Humblet-Baron; Laurence Seidel; Rémi Cheynier; Evelyne Willems; André Gothot; Jean-François Vanbellinghen; Vincent Geenen; Brenda M. Sandmaier; Rainer Storb; Yves Beguin; Frédéric Baron

Background Long-term immune recovery in older patients given hematopoietic cell transplantation after non-myeloablative conditioning remains poorly understood. This prompted us to investigate long-term lymphocyte reconstitution and thymic function in 80 patients given allogeneic peripheral blood stem cells after non-myeloablative conditioning. Design and Methods Median age at transplant was 57 years (range 10–71). Conditioning regimen consisted of 2 Gy total body irradiation (TBI) with (n=46) or without (n=20) added fludarabine, 4 Gy TBI with fludarabine (n=6), or cyclophosphamide plus fludarabine (n=8). Stem cell sources were unmanipulated (n=56), CD8-depleted (n=19), or CD34-selected (n=5) peripheral blood stem cells. Immune recovery was assessed by signal-joint T-cell receptor excision circle quantification and flow cytometry. Results Signal-joint T-cell receptor excision circle levels increased from day 100 to one and two years after transplantation in patients under 50 years of age (n=23; P=0.02 and P=0.04, respectively), and in those aged 51–60 years (n=35; P=0.17 and P=0.06, respectively), but not in patients aged over 60 (n=22; P=0.3 and P=0.3, respectively). Similarly, CD4+CD45RA+ (naïve) T-cell counts increased from day 100 to one and two years after transplantation in patients aged 50 years and under 50 (P=0.002 and P=0.02, respectively), and in those aged 51–60 (P=0.4 and P=0.001, respectively), but less so in patients aged over 60 (P=0.3 and P=0.06, respectively). In multivariate analyses, older patient age (P<0.001), extensive chronic GVHD (P<0.001), and prior (resolved) extensive chronic graft-versus-host disease (P=0.008) were associated with low signal-joint T-cell receptor excision circle levels one year or more after HCT. Conclusions In summary, our data suggest that thymic neo-generation of T cells occurred from day 100 onwards in patients under 60 while signal-joint T-cell receptor excision circle levels remained low for patients aged over 60. Further, chronic graft-versus-host disease had a dramatic impact on thymic function, as observed previously in patients given grafts after myeloablative conditioning.


Endocrine-related Cancer | 2015

Clinical and genetic characterization of pituitary gigantism: an international collaborative study in 208 patients

Liliya Rostomyan; Adrian Daly; Patrick Petrossians; Emil Nachev; Anurag Lila; Anne Lise Lecoq; Beatriz Lecumberri; Giampaolo Trivellin; Roberto Salvatori; Andreas G. Moraitis; Ian Holdaway; Dianne J. Kranenburg-Van Klaveren; Maria Chiara Zatelli; Nuria Palacios; Cécile Nozières; Margaret Zacharin; Tapani Ebeling; Marja Ojaniemi; Liudmila Rozhinskaya; Elisa Verrua; Marie Lise Jaffrain-Rea; Silvia Filipponi; Daria Gusakova; Vyacheslav Pronin; Jérôme Bertherat; Zhanna Belaya; Irena Ilovayskaya; Mona Sahnoun-Fathallah; Caroline Sievers; Günter K. Stalla

Despite being a classical growth disorder, pituitary gigantism has not been studied previously in a standardized way. We performed a retrospective, multicenter, international study to characterize a large series of pituitary gigantism patients. We included 208 patients (163 males; 78.4%) with growth hormone excess and a current/previous abnormal growth velocity for age or final height >2 s.d. above country normal means. The median onset of rapid growth was 13 years and occurred significantly earlier in females than in males; pituitary adenomas were diagnosed earlier in females than males (15.8 vs 21.5 years respectively). Adenomas were ≥10 mm (i.e., macroadenomas) in 84%, of which extrasellar extension occurred in 77% and invasion in 54%. GH/IGF1 control was achieved in 39% during long-term follow-up. Final height was greater in younger onset patients, with larger tumors and higher GH levels. Later disease control was associated with a greater difference from mid-parental height (r=0.23, P=0.02). AIP mutations occurred in 29%; microduplication at Xq26.3 - X-linked acrogigantism (X-LAG) - occurred in two familial isolated pituitary adenoma kindreds and in ten sporadic patients. Tumor size was not different in X-LAG, AIP mutated and genetically negative patient groups. AIP-mutated and X-LAG patients were significantly younger at onset and diagnosis, but disease control was worse in genetically negative cases. Pituitary gigantism patients are characterized by male predominance and large tumors that are difficult to control. Treatment delay increases final height and symptom burden. AIP mutations and X-LAG explain many cases, but no genetic etiology is seen in >50% of cases.


Endocrine-related Cancer | 2016

Somatic mosaicism underlies X-linked acrogigantism syndrome in sporadic male subjects

Adrian Daly; Bo Yuan; Frédéric Fina; Jean-Hubert Caberg; Giampaolo Trivellin; Liliya Rostomyan; Wouter W. de Herder; Luciana A. Naves; D Metzger; Thomas Cuny; Wolfgang Rabl; Nalini S. Shah; Marie-Lise Jaffrain-Rea; Maria Chiara Zatelli; Fabio R. Faucz; Emilie Castermans; Isabelle Nanni-Metellus; Maya Lodish; Ammar Muhammad; Leonor Palmeira; Iulia Potorac; Giovanna Mantovani; Sebastian Neggers; M. Klein; Anne Barlier; Pengfei Liu; L’Houcine Ouafik; Vincent Bours; James R. Lupski; Constantine A. Stratakis

Somatic mosaicism has been implicated as a causative mechanism in a number of genetic and genomic disorders. X-linked acrogigantism (XLAG) syndrome is a recently characterized genomic form of pediatric gigantism due to aggressive pituitary tumors that is caused by submicroscopic chromosome Xq26.3 duplications that include GPR101 We studied XLAG syndrome patients (n= 18) to determine if somatic mosaicism contributed to the genomic pathophysiology. Eighteen subjects with XLAG syndrome caused by Xq26.3 duplications were identified using high-definition array comparative genomic hybridization (HD-aCGH). We noted that males with XLAG had a decreased log2ratio (LR) compared with expected values, suggesting potential mosaicism, whereas females showed no such decrease. Compared with familial male XLAG cases, sporadic males had more marked evidence for mosaicism, with levels of Xq26.3 duplication between 16.1 and 53.8%. These characteristics were replicated using a novel, personalized breakpoint junction-specific quantification droplet digital polymerase chain reaction (ddPCR) technique. Using a separate ddPCR technique, we studied the feasibility of identifying XLAG syndrome cases in a distinct patient population of 64 unrelated subjects with acromegaly/gigantism, and identified one female gigantism patient who had had increased copy number variation (CNV) threshold for GPR101 that was subsequently diagnosed as having XLAG syndrome on HD-aCGH. Employing a combination of HD-aCGH and novel ddPCR approaches, we have demonstrated, for the first time, that XLAG syndrome can be caused by variable degrees of somatic mosaicism for duplications at chromosome Xq26.3. Somatic mosaicism was shown to occur in sporadic males but not in females with XLAG syndrome, although the clinical characteristics of the disease were similarly severe in both sexes.


Haematologica | 2008

Evidence for neo-generation of T cells by the thymus after non-myeloablative conditioning

Emilie Castermans; Frédéric Baron; Evelyne Willems; Nicole Schaaf-Lafontaine; Nathalie Meuris; André Gothot; Jean-François Vanbellighen; Christian Herens; Laurence Seidel; Vincent Geenen; Rémi Cheynier; Yves Beguin

In patients given allogeneic stem cell transplantation with non-myeloablative conditioning, immune recovery is initially driven by peripheral expansion of the graft-contained mature T cell, while T-cell neo-generation by the thymus plays an important role in long-term immune reconstitution. Background Background and objective. We investigated immune recovery in 50 patients given either unmanipulated or CD8-depleted allogeneic peripheral blood stem cells after non-myeloablative conditioning. Design and Methods Fifty patients were randomized to receive either CD8-depleted (n=22) or non-manipulated (n=28) peripheral blood stem cells. The median patients age was 57 (range 36–69) years. The conditioning regimen consisted of 2 Gy total body irradiation with or without added fludarabine. Twenty patients received grafts from related donors, 14 from 10/10 HLA-allele matched unrelated donors, and 16 from HLA-mismatched unrelated donors. Graft-versus-host disease pro-phylaxis consisted of mycophenolate mofetil and cyclosporine. Immune recovery during the first year after hematopoietic cell transplantation was assessed by flow cytometry phenotyping, analyses of the diversity of the TCRBV repertoire, and quantification of signal-joint T-cell receptor excision circles (sjTREC). Results CD8-depletion of the graft reduced the recovery of CD8+ T-cell counts in the first 6 months following transplantation (p<0.0001) but had no significant impact on the restoration of other T-cell subsets. Both sjTREC concentration and CD3+ T-cell counts increased significantly between day 100 and 365 (p=0.010 and p=0.0488, respectively) demonstrating neo-production of T cells by the thymus. Factors associated with high sjTREC concentration 1 year after transplantation included an HLA-matched unrelated donor (p=0.029), a high content of T cells in the graft (p=0.002), and the absence of chronic graft-versus-host disease (p<0.0001). Conclusions Our data suggest that while immune recovery is mainly driven by peripheral expansion of the graft-contained mature T cells during the first months after non-myeloablative transplantation, T-cell neo-generation by the thymus plays an important role in long term immune reconstitution in transplanted patients.


Transplantation | 2003

T-CELL RECONSTITUTION AFTER UNMANIPULATED, CD8- DEPLETED OR CD34-SELECTED NONMYELOABLATIVE PERIPHERAL BLOOD STEM-CELL TRANSPLANTATION

Frédéric Baron; Nicole Schaaf-Lafontaine; Stéphanie Humblet-Baron; Nathalie Meuris; Emilie Castermans; Etienne Baudoux; Pascale Frere; Vincent Bours; Georges Fillet; Yves Beguin

Background. We have previously shown that CD8 depletion or CD34 selection of peripheral blood stem cells (PBSC) reduced the incidence of acute graft-versus-host disease (GvHD) after nonmyeloablative stem-cell transplantation (NMSCT). In this study, we analyze the effect of CD8 depletion or CD34 selection of the graft on early T-cell reconstitution. Methods. Nonmyeloablative conditioning regimen consisted in 2 Gy total-body irradiation (TBI) alone, 2 Gy TBI and fludarabine, or cyclophosphamide and fludarabine. Patients 1 to 18 received unmanipulated PBSC, patients 19 to 29 CD8-depleted PBSC, and patients 30 to 35 CD34-selected PBSC. Results. T-cell counts, and particularly CD4+ and CD4CD45RA+ counts, remained low the first 6 months after nonmyeloablative stem-cell transplantation (NMSCT) in all patients. CD34 selection (P <0.0001) but not CD8 depletion of PBSC significantly decreased T-cell chimerism. Donor T-cell count was similar in unmanipulated compared with CD8-depleted PBSC recipients but was significantly lower in CD34-selected PBSC recipients (P =0.0012). T cells of recipient origin remained stable over time in unmanipulated and CD8-depleted PBSC patients but expanded in some CD34-selected PBSC recipients between day 28 and 100 after transplant. Moreover, whereas CD8 depletion only decreased CD8+ counts (P <0.047), CD34 selection reduced CD3+(P <0.001), CD8+(P <0.016), CD4+ (P <0.001), and CD4+CD45RA+ (P <0.001) cell counts. T-cell repertoire was restricted in all patients on day 100 after hematopoietic stem-cell transplantation but was even more limited after CD34 selection (P =0.002). Conclusions. Despite of the persistence of a significant number of T cells of recipient origin, T-cell counts were low the first 6 months after NMSCT. Moreover, contrary with CD8 depletion of the graft that only affects CD8+ lymphocyte counts, CD34 selection dramatically decreased both CD8 and CD4 counts.


Endocrine-related Cancer | 2016

GHRH excess and blockade in X-LAG syndrome

Adrian Daly; Philippe A. Lysy; Céline Desfilles; Liliya Rostomyan; Amira Mohamed; Jean-Hubert Caberg; Veronique Raverot; Emilie Castermans; Etienne Marbaix; Dominique Maiter; Chloé Brunelle; Giampaolo Trivellin; Constantine A. Stratakis; Vincent Bours; Christian Raftopoulos; Véronique Beauloye; Anne Barlier; Albert Beckers

X-linked acrogigantism (X-LAG) syndrome is a newly described form of inheritable pituitary gigantism that begins in early childhood and is usually associated with markedly elevated GH and prolactin secretion by mixed pituitary adenomas/hyperplasia. Microduplications on chromosome Xq26.3 including the GPR101 gene cause X-LAG syndrome. In individual cases random GHRH levels have been elevated. We performed a series of hormonal profiles in a young female sporadic X-LAG syndrome patient and subsequently undertook in vitro studies of primary pituitary tumor culture following neurosurgical resection. The patient demonstrated consistently elevated circulating GHRH levels throughout preoperative testing, which was accompanied by marked GH and prolactin hypersecretion; GH demonstrated a paradoxical increase following TRH administration. In vitro, the pituitary cells showed baseline GH and prolactin release that was further stimulated by GHRH administration. Co-incubation with GHRH and the GHRH receptor antagonist, acetyl-(d-Arg(2))-GHRH (1-29) amide, blocked the GHRH-induced GH stimulation; the GHRH receptor antagonist alone significantly reduced GH release. Pasireotide, but not octreotide, inhibited GH secretion. A ghrelin receptor agonist and an inverse agonist led to modest, statistically significant increases and decreases in GH secretion, respectively. GHRH hypersecretion can accompany the pituitary abnormalities seen in X-LAG syndrome. These data suggest that the pathology of X-LAG syndrome may include hypothalamic dysregulation of GHRH secretion, which is in keeping with localization of GPR101 in the hypothalamus. Therapeutic blockade of GHRH secretion could represent a way to target the marked hormonal hypersecretion and overgrowth that characterizes X-LAG syndrome.


Stem Cells | 2006

Despite Inhibition of Hematopoietic Progenitor Cell Growth In Vitro, the Tyrosine Kinase Inhibitor Imatinib Does Not Impair Engraftment of Human CD133+ Cells into NOD/SCIDβ2mNull Mice

Laurence Pirson; Frédéric Baron; Nathalie Meuris; Olivier Giet; Emilie Castermans; Roland Greimers; Ivano Di Stefano; André Gothot; Yves Beguin

There is potential interest for combining allogeneic hematopoietic cell transplantation (HCT), and particularly allogeneic HCT with a nonmyeloablative regimen, to the tyrosine kinase inhibitor imatinib (Glivec; Novartis, Basel, Switzerland, http://www.novartis.com) in order to maximize anti‐leukemic activity against Philadelphia chromosome‐positive leukemias. However, because imatinib inhibits c‐kit, the stem cell factor receptor, it could interfere with bone marrow engraftment. In this study, we examined the impact of imatinib on normal progenitor cell function. Imatinib decreased the colony‐forming capacity of mobilized peripheral blood human CD133+ cells but not that of long‐term culture‐initiating cells. Imatinib also decreased the proliferation of cytokine‐stimulated CD133+ cells but did not induce apoptosis of these cells. Expression of very late antigen (VLA)‐4, VLA‐5, and CXCR4 of CD133+ cells was not modified by imatinib, but imatinib decreased the ability of CD133+ cells to migrate. Finally, imatinib did not decrease engraftment of CD133+ cells into irradiated nonobese diabetic/severe combined immunodeficient/β2mnull mice conditioned with 3 or 1 Gy total body irradiation. In summary, our results suggest that, despite inhibition of hematopoietic progenitor cell growth in vitro, imatinib does not interfere with hematopoietic stem cell engraftment.


Familial Cancer | 2014

Deletion of exons 1-3 of the MEN1 gene in a large Italian family causes the loss of menin expression.

Maria Chiara Zatelli; Federico Tagliati; Mauro Di Ruvo; Emilie Castermans; Luigi Cavazzini; Adrian Daly; Maria Rosaria Ambrosio; Albert Beckers; Ettore C. degli Uberti

Multiple endocrine neoplasia type 1 (MEN1) syndrome is an autosomal dominant disease, characterized by parathyroid adenomas, endocrine gastroenteropancreatic tumors and pituitary adenomas, due to inactivating mutations of the MEN1 gene (chromosome 11q13). MEN1 mutations are mainly represented by nonsense, deletions/insertions, splice site or missense mutations that can be detected by direct sequencing of genomic DNA. However, MEN1 patients with large heterozygous deletions may escape classical genetic screening and may be misidentified as phenocopies, thereby hindering proper clinical surveillance. We employed a real-time polymerase chain reaction application, the TaqMan copy number variation assay, to evaluate a family in which we failed to identify an MEN1 mutation by direct sequencing, despite a clear clinical diagnosis of MEN1 syndrome. Using the TaqMan copy number variation assay we identified a large deletion of the MEN1 gene involving exons 1 and 2, in three affected family members, but not in the other nine family members that were to date clinically unaffected. The same genetic alteration was not found in a group of ten unaffected subjects, without family history of endocrine tumors. The MEN1 deletion was further confirmed by multiplex ligation-dependent probe amplification, which showed the deletion extended from exon 1 to exon 3. This new approach allowed us to correctly genetically diagnose three clinical MEN1 patients that were previously considered as MEN1 phenocopies. More importantly, we excluded the presence of genetic alterations in the unaffected family members. These results underline the importance of using a variety of available biotechnology approaches when pursuing a genetic diagnosis in a clinically suggestive setting of inherited endocrine cancer.

Collaboration


Dive into the Emilie Castermans's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giampaolo Trivellin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nalini S. Shah

King Edward Memorial Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge