Network


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

Hotspot


Dive into the research topics where Heleen Roose is active.

Publication


Featured researches published by Heleen Roose.


Development | 2017

Development of organoids from mouse and human endometrium showing endometrial epithelium physiology and long-term expandability

Matteo Giovanni Maria Boretto; Benoit Cox; Manuel Noben; Nikolai Hendriks; Amelie Fassbender; Heleen Roose; Frédéric Amant; Dirk Timmerman; Carla Tomassetti; Arne Vanhie; Christel Meuleman; Marc Ferrante; Hugo Vankelecom

The endometrium, which is of crucial importance for reproduction, undergoes dynamic cyclic tissue remodeling. Knowledge of its molecular and cellular regulation is poor, primarily owing to a lack of study models. Here, we have established a novel and promising organoid model from both mouse and human endometrium. Dissociated endometrial tissue, embedded in Matrigel under WNT-activating conditions, swiftly formed organoid structures that showed long-term expansion capacity, and reproduced the molecular and histological phenotype of the tissues epithelium. The supplemented WNT level determined the type of mouse endometrial organoids obtained: high WNT yielded cystic organoids displaying a more differentiated phenotype than the dense organoids obtained in low WNT. The organoids phenocopied physiological responses of endometrial epithelium to hormones, including increased cell proliferation under estrogen and maturation upon progesterone. Moreover, the human endometrial organoids replicated the menstrual cycle under hormonal treatment at both the morpho-histological and molecular levels. Together, we established an organoid culture system for endometrium, reproducing tissue epithelium physiology and allowing long-term expansion. This novel model provides a powerful tool for studying mechanisms underlying the biology as well as the pathology of this key reproductive organ. Summary: Human endometrium organoids replicate the menstrual cycle in response to hormone treatment, while mouse endometrium organoid type is determined by variations in WNT.


Endocrinology | 2016

Regeneration in the pituitary after cell-ablation injury: time-related aspects and molecular analysis.

Christophe Willems; Qiuli Fu; Heleen Roose; Freya Mertens; Benoit Cox; Jianghai Chen; Hugo Vankelecom

We recently showed that the mouse pituitary holds regenerative competence. Young-adult GHCre/iDTR mice, expressing diphtheria toxin (DT) receptor in GH-producing cells, regenerate the GH(+) cells, as ablated by 3-day DT treatment (3DT), up to 60% after 5 months. The pituitarys stem cells participate in this restoration process. Here, we characterized this regenerative capacity in relation to age and recovery period and started to search for underlying molecular mechanisms. Extending the recovery period (up to 19 mo) does not result in higher regeneration levels. In addition, the regenerative competence disappears at older age, coinciding with a reduction in pituitary stem cell number and fitness. Surprisingly, prolonging DT treatment of young-adult mice to 10 days (10DT) completely blocks the regeneration, although the stem cell compartment still reacts by promptly expanding, and retains in vitro stem cell functionality. To obtain a first broad view on molecular grounds underlying reparative capacity and/or failure, the stem cell-clustering side population was analyzed by whole-genome expression analysis. A number of stemness factors and components of embryonic, epithelial-mesenchymal transition, growth factor and Hippo pathways are higher expressed in the stem cell-clustering side population of the regenerating pituitary (after 3DT) when compared with the basal gland and to the nonregenerating pituitary (after 10DT). Together, the regenerative capacity of the pituitary is limited both in age-related terms and final efficacy, and appears to rely on stem cell-associated pathway activation. Dissection of the molecular profiles may eventually identify targets to induce or boost regeneration in situations of (injury-related) pituitary deficiency.


Autophagy | 2017

An autophagy-driven pathway of ATP secretion supports the aggressive phenotype of BRAFV600E inhibitor-resistant metastatic melanoma cells

Shaun Martin; Aleksandra M. Dudek-Peric; Abhishek D. Garg; Heleen Roose; Seyma Demirsoy; Sofie Van Eygen; Freya Mertens; Peter Vangheluwe; Hugo Vankelecom; Patrizia Agostinis

ABSTRACT The ingrained capacity of melanoma cells to rapidly evolve toward an aggressive phenotype is manifested by their increased ability to develop drug-resistance, evident in the case of vemurafenib, a therapeutic-agent targeting BRAFV600E. Previous studies indicated a tight correlation between heightened melanoma-associated macroautophagy/autophagy and acquired Vemurafenib resistance. However, how this vesicular trafficking pathway supports Vemurafenib resistance remains unclear. Here, using isogenic human and murine melanoma cell lines of Vemurafenib-resistant and patient-derived melanoma cells with primary resistance to the BRAFV600E inhibitor, we found that the enhanced migration and invasion of the resistant melanoma cells correlated with an enhanced autophagic capacity and autophagosome-mediated secretion of ATP. Extracellular ATP (eATP) was instrumental for the invasive phenotype and the expansion of a subset of Vemurafenib-resistant melanoma cells. Compromising the heightened autophagy in these BRAFV600E inhibitor-resistant melanoma cells through the knockdown of different autophagy genes (ATG5, ATG7, ULK1), reduced their invasive and eATP-secreting capacity. Furthermore, eATP promoted the aggressive nature of the BRAFV600E inhibitor-resistant melanoma cells by signaling through the purinergic receptor P2RX7. This autophagy-propelled eATP-dependent autocrine-paracrine pathway supported the maintenance and expansion of a drug-resistant melanoma phenotype. In conclusion, we have identified an autophagy-driven response that relies on the secretion of ATP to drive P2RX7-based migration and expansion of the Vemurafenib-resistant phenotype. This emphasizes the potential of targeting autophagy in the treatment and management of metastatic melanoma.


Scientific Reports | 2017

Major depletion of SOX2 + stem cells in the adult pituitary is not restored which does not affect hormonal cell homeostasis and remodelling

Heleen Roose; Benoit Cox; Matteo Giovanni Maria Boretto; Conny Gysemans; Annelies Vennekens; Hugo Vankelecom

The pituitary gland contains SOX2-expressing stem cells. However, their functional significance remains largely unmapped. We investigated their importance by depleting SOX2+ cells through diphtheria toxin (DT)-mediated ablation. DT treatment of adult Sox2CreERT2/+;R26iDTR/+ mice (after tamoxifen-induced expression of DT receptor in SOX2+ cells) resulted in 80% obliteration of SOX2+ cells in the endocrine pituitary, coinciding with reduced pituisphere-forming activity. Counterintuitively for a stem cell population, the SOX2+ cell compartment did not repopulate. Considering the more active phenotype of the stem cells during early-postnatal pituitary maturation, SOX2+ cell ablation was also performed in 4- and 1-week-old animals. Ablation grade diminished with decreasing age and was accompanied by a proliferative reaction of the SOX2+ cells, suggesting a rescue attempt. Despite this activation, SOX2+ cells did also not recover. Finally, the major SOX2+ cell depletion in adult mice did not affect the homeostatic maintenance of pituitary hormonal cell populations, nor the corticotrope remodelling response to adrenalectomy challenge. Taken together, our study shows that pituitary SOX2+ fail to regenerate after major depletion which does not affect adult endocrine cell homeostasis and remodelling. Thus, pituitary SOX2+ cells may constitute a copious stem cell reserve or may have other critical role(s) still to be clearly defined.


Frontiers in Endocrinology | 2017

The Stem Cell Connection of Pituitary Tumors

Hugo Vankelecom; Heleen Roose

Tumors in the pituitary gland are typically benign but cause serious morbidity due to compression of neighboring structures and hormonal disruptions. Overall, therapy efficiency remains suboptimal with negative impact on health and comfort of life, including considerable risk of therapy resistance and tumor recurrence. To date, little is known on the pathogenesis of pituitary tumors. Stem cells may represent important forces in this process. The pituitary tumors may contain a driving tumor stem cell population while the resident tissue stem cells may be directly or indirectly linked to tumor development and growth. Here, we will briefly summarize recent studies that afforded a glance behind the scenes of this stem cell connection. A better knowledge of the mechanisms underlying pituitary tumorigenesis is essential to identify more efficacious treatment modalities and improve clinical management.


Journal of Endocrinology | 2017

Pituitary stem cell regulation: who is pulling the strings?

Benoit Cox; Heleen Roose; Annelies Vennekens; Hugo Vankelecom


Society for Endocrinology BES 2016 | 2016

Pituitary tumors contain a side population with tumor stem cell-associated characteristics

Freya Mertens; Lies Gremeaux; Jianghai Chen; Qiuli Fu; Christophe Williems; Heleen Roose; Olivier Govaere; Tania Roskams; Carolina Cristina; Damasia Becu-Villalobos; Mark Jorissen; Vincent Vander Poorten; Marie Bex; Loon Johannes van; Hugo Vankelecom


Archive | 2015

Pituitary hormonal cells from human pluripotent stem cells

Hugo Vankelecom; Benoit Cox; An-Katrien Theunis; Freya Mertens; Heleen Roose; Christophe Willems


Archive | 2015

Endometrial cancer: in search of cancer stem cells and disease modeling in a dish

Matteo Giovanni Maria Boretto; Benoit Cox; Freya Mertens; Heleen Roose; Christophe Willems; Debby Thomas; Els Hermans; Frédéric Amant; Hugo Vankelecom


Archive | 2015

Identification of a candidate ‘cancer stem cell’ population in human endometrial cancer and xenograft tumors

Matteo Giovanni Maria Boretto; Benoit Cox; Freya Mertens; Heleen Roose; Christophe Willems; Debby Thomas; Els Hermans; Frédéric Amant; Hugo Vankelecom

Collaboration


Dive into the Heleen Roose's collaboration.

Top Co-Authors

Avatar

Hugo Vankelecom

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Benoit Cox

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Freya Mertens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Christophe Willems

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annelies Vennekens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Frédéric Amant

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jianghai Chen

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Qiuli Fu

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Debby Thomas

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge