Network


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

Hotspot


Dive into the research topics where Loic P. Deleyrolle is active.

Publication


Featured researches published by Loic P. Deleyrolle.


Stem Cells | 2008

Enumeration of neural stem and progenitor cells in the neural colony-forming cell assay.

Sharon A. Louis; Rodney L. Rietze; Loic P. Deleyrolle; Ravenska Wagey; Terry Thomas; Allen C. Eaves; Brent A. Reynolds

Advancement in our understanding of the biology of adult stem cells and their therapeutic potential relies heavily on meaningful functional assays that can identify and measure stem cell activity in vivo and in vitro. In the mammalian nervous system, neural stem cells (NSCs) are often studied using a culture system referred to as the neurosphere assay. We previously challenged a central tenet of this assay, that all neurospheres are derived from a NSC, and provided evidence that it overestimates NSC frequency, rendering it inappropriate for quantitation of NSC frequency in relation to NSC regulation. Here we report the development and validation of the neural colony‐forming cell assay (NCFCA), which discriminates stem from progenitor cells on the basis of their proliferative potential. We anticipate that the NCFCA will provide additional clarity in discerning the regulation of NSCs, thereby facilitating further advances in the promising application of NSCs for therapeutic use.


Embo Molecular Medicine | 2013

The ZEB1 pathway links glioblastoma initiation, invasion and chemoresistance

Florian A. Siebzehnrubl; Daniel J. Silver; Bugra Tugertimur; Loic P. Deleyrolle; Dorit Siebzehnrubl; Matthew R. Sarkisian; Kelly G. Devers; Antony Yachnis; Marius D. Kupper; Daniel Neal; Nancy H. Nabilsi; Michael P. Kladde; Oleg Suslov; Simone Brabletz; Thomas Brabletz; Brent A. Reynolds; Dennis A. Steindler

Glioblastoma remains one of the most lethal types of cancer, and is the most common brain tumour in adults. In particular, tumour recurrence after surgical resection and radiation invariably occurs regardless of aggressive chemotherapy. Here, we provide evidence that the transcription factor ZEB1 (zinc finger E‐box binding homeobox 1) exerts simultaneous influence over invasion, chemoresistance and tumourigenesis in glioblastoma. ZEB1 is preferentially expressed in invasive glioblastoma cells, where the ZEB1‐miR‐200 feedback loop interconnects these processes through the downstream effectors ROBO1, c‐MYB and MGMT. Moreover, ZEB1 expression in glioblastoma patients is predictive of shorter survival and poor Temozolomide response. Our findings indicate that this regulator of epithelial‐mesenchymal transition orchestrates key features of cancer stem cells in malignant glioma and identify ROBO1, OLIG2, CD133 and MGMT as novel targets of the ZEB1 pathway. Thus, ZEB1 is an important candidate molecule for glioblastoma recurrence, a marker of invasive tumour cells and a potential therapeutic target, along with its downstream effectors.


Methods of Molecular Biology | 2009

Isolation, expansion, and differentiation of adult Mammalian neural stem and progenitor cells using the neurosphere assay.

Loic P. Deleyrolle; Brent A. Reynolds

During development and continuing into adulthood, stem cells function as a reservoir of undifferentiated cell types, whose role is to support cell genesis in several tissues and organs. In the adult, they play an essential homeostatic role by replacing differentiated cells that are lost due to physiological turnover, injury, or disease. The discovery of such cells in the adult mammalian central nervous system (CNS), an organ traditionally thought to have little or no regenerative capacity, has opened the door to the possibility of designing innovative regenerative therapeutics, an unexpected concept in neurobiology 15 years ago. In 1992, to detect precursor cells in the adult brain, we employed a serum-free culture system whereby the majority of primary differentiated CNS cells did not survive but a small population of EGF-responsive cells were maintained in an undifferentiated state and proliferated to form clusters, called neurospheres (Reynolds and Weiss, Science 255:1707-1710, 1992). These neurospheres could be (a) dissociated to form numerous secondary spheres or (b) induced to differentiate, generating the three major cell types of the CNS. This chapter outlines the adult mammalian NSC culture methodology and provides technical details of the neurosphere assay to achieve reproducible cultures.


Brain | 2011

Evidence for label-retaining tumour-initiating cells in human glioblastoma

Loic P. Deleyrolle; Angus Harding; Kathleen Cato; Florian A. Siebzehnrubl; Maryam Rahman; Hassan Azari; Sarah Olson; Brian Gabrielli; Geoffrey W. Osborne; Angelo L. Vescovi; Brent A. Reynolds

Individual tumour cells display diverse functional behaviours in terms of proliferation rate, cell-cell interactions, metastatic potential and sensitivity to therapy. Moreover, sequencing studies have demonstrated surprising levels of genetic diversity between individual patient tumours of the same type. Tumour heterogeneity presents a significant therapeutic challenge as diverse cell types within a tumour can respond differently to therapies, and inter-patient heterogeneity may prevent the development of general treatments for cancer. One strategy that may help overcome tumour heterogeneity is the identification of tumour sub-populations that drive specific disease pathologies for the development of therapies targeting these clinically relevant sub-populations. Here, we have identified a dye-retaining brain tumour population that displays all the hallmarks of a tumour-initiating sub-population. Using a limiting dilution transplantation assay in immunocompromised mice, label-retaining brain tumour cells display elevated tumour-initiation properties relative to the bulk population. Importantly, tumours generated from these label-retaining cells exhibit all the pathological features of the primary disease. Together, these findings confirm dye-retaining brain tumour cells exhibit tumour-initiation ability and are therefore viable targets for the development of therapeutics targeting this sub-population.


Neurosurgery | 2011

The Cancer Stem Cell Hypothesis: Failures and Pitfalls

Maryam Rahman; Loic P. Deleyrolle; Vinata Vedam-Mai; Hassan Azari; Muhammad M. Abd-El-Barr; Brent A. Reynolds

Based on the clonal evolution model and the assumption that the vast majority of tumor cells are able to propagate and drive tumor growth, the goal of cancer treatment has traditionally been to kill all cancerous cells. This theory has been challenged recently by the cancer stem cell (CSC) hypothesis, that a rare population of tumor cells, with stem cell characteristics, is responsible for tumor growth, resistance, and recurrence. Evidence for putative CSCs has been described in blood, breast, lung, prostate, colon, liver, pancreas, and brain. This new hypothesis would propose that indiscriminate killing of cancer cells would not be as effective as selective targeting of the cells that are driving long-term growth (ie, the CSCs) and that treatment failure is often the result of CSCs escaping traditional therapies. The CSC hypothesis has gained a great deal of attention because of the identification of a new target that may be responsible for poor outcomes of many aggressive cancers, including malignant glioma. As attractive as this hypothesis sounds, especially when applied to tumors that respond poorly to current treatments, we will argue in this article that the proposal of a stemlike cell that initiates and drives solid tissue cancer growth and is responsible for therapeutic failure is far from proven. We will present the point of view that for most advanced solid tissue cancers such as glioblastoma multiforme, targeting a putative rare CSC population will have little effect on patient outcomes. This review will cover problems with the CSC hypothesis, including applicability of the hierarchical model, inconsistencies with xenotransplantation data, and nonspecificity of CSC markers.


PLOS ONE | 2011

Determination of Somatic and Cancer Stem Cell Self-Renewing Symmetric Division Rate Using Sphere Assays

Loic P. Deleyrolle; Geoffery Ericksson; Brian J. Morrison; J. Alejandro Lopez; Kevin Burrage; Pamela Burrage; Angelo L. Vescovi; Rodney L. Rietze; Brent A. Reynolds

Representing a renewable source for cell replacement, neural stem cells have received substantial attention in recent years. The neurosphere assay represents a method to detect the presence of neural stem cells, however owing to a deficiency of specific and definitive markers to identify them, their quantification and the rate they expand is still indefinite. Here we propose a mathematical interpretation of the neurosphere assay allowing actual measurement of neural stem cell symmetric division frequency. The algorithm of the modeling demonstrates a direct correlation between the overall cell fold expansion over time measured in the sphere assay and the rate stem cells expand via symmetric division. The model offers a methodology to evaluate specifically the effect of diseases and treatments on neural stem cell activity and function. Not only providing new insights in the evaluation of the kinetic features of neural stem cells, our modeling further contemplates cancer biology as cancer stem-like cells have been suggested to maintain tumor growth as somatic stem cells maintain tissue homeostasis. Indeed, tumor stem cells resistance to therapy makes these cells a necessary target for effective treatment. The neurosphere assay mathematical model presented here allows the assessment of the rate malignant stem-like cells expand via symmetric division and the evaluation of the effects of therapeutics on the self-renewal and proliferative activity of this clinically relevant population that drive tumor growth and recurrence.


Glia | 2011

The origins of glioma: E Pluribus Unum?

Florian A. Siebzehnrubl; Brent A. Reynolds; Angelo L. Vescovi; Dennis A. Steindler; Loic P. Deleyrolle

Malignant glioma is among of the most devastating, and least curable, types of cancer. Since the re‐emergence of the cancer stem cell hypothesis, much progress has been made towards elucidating the cellular origin of these tumors. The hypothesis that tumors are hierarchically organized, with a cancer stem cell at the top that shares defining features with somatic stem cells and provides therapeutic refractoriness properties, has put adult stem cells into the limelight as prime suspect for malignant glioma. Much confusion still exists, though, as to the particular cell type and processes that lead to oncogenic transformation. In this review, we will discuss recent developments and novel hypotheses regarding the origin of malignant gliomas, especially glioblastoma. In particular, we argue that glioblastoma is the result of different pathways originating in multiple sources that all ultimately converge in the same disease. Further attention is devoted to potential scenarios leading to transformation of different stem/progenitor cell types of the brain, and the probability and relevance of these scenarios for malignant tumorigenesis.


Cancer Research | 2011

YB-1 Bridges Neural Stem Cells and Brain Tumor–Initiating Cells via Its Roles in Differentiation and Cell Growth

Abbas Fotovati; Samah Abu-Ali; Pei-Shan Wang; Loic P. Deleyrolle; Cathy Lee; Joanna Triscott; James Y. Chen; Sonia Franciosi; Yasuhiro Nakamura; Yasuo Sugita; Takeshi Uchiumi; Michihiko Kuwano; Blair R. Leavitt; Sheila K. Singh; Alexa Jury; Chris Jones; Hiroaki Wakimoto; Brent A. Reynolds; Catherine J. Pallen; Sandra E. Dunn

The Y-box binding protein 1 (YB-1) is upregulated in many human malignancies including glioblastoma (GBM). It is also essential for normal brain development, suggesting that YB-1 is part of a neural stem cell (NSC) network. Here, we show that YB-1 was highly expressed in the subventricular zone (SVZ) of mouse fetal brain tissues but not in terminally differentiated primary astrocytes. Conversely, YB-1 knockout mice had reduced Sox-2, nestin, and musashi-1 expression in the SVZ. Although primary murine neurospheres were rich in YB-1, its expression was lost during glial differentiation. Glial tumors often express NSC markers and tend to loose the cellular control that governs differentiation; therefore, we addressed whether YB-1 served a similar role in cancer cells. YB-1, Sox-2, musashi-1, Bmi-1, and nestin are coordinately expressed in SF188 cells and 9/9 GBM patient-derived primary brain tumor-initiating cells (BTIC). Silencing YB-1 with siRNA attenuated the expression of these NSC markers, reduced neurosphere growth, and triggered differentiation via coordinate loss of GSK3-β. Furthermore, differentiation of BTIC with 1% serum or bone morphogenetic protein-4 suppressed YB-1 protein expression. Likewise, YB-1 expression was lost during differentiation of normal human NSCs. Consistent with these observations, YB-1 expression increased with tumor grade (n = 49 cases). YB-1 was also coexpressed with Bmi-1 (Spearmans 0.80, P > 0.001) and Sox-2 (Spearmans 0.66, P > 0.001) based on the analysis of 282 cases of high-grade gliomas. These proteins were highly expressed in 10/15 (67%) of GBM patients that subsequently relapsed. In conclusion, YB-1 correlatively expresses with NSC markers where it functions to promote cell growth and inhibit differentiation.


PLOS ONE | 2011

Purification of immature neuronal cells from neural stem cell progeny

Hassan Azari; Geoffrey W. Osborne; Takahiro Yasuda; Mohammad G. Golmohammadi; Maryam Rahman; Loic P. Deleyrolle; Ebrahim Esfandiari; David J. Adams; Björn Scheffler; Dennis A. Steindler; Brent A. Reynolds

Large-scale proliferation and multi-lineage differentiation capabilities make neural stem cells (NSCs) a promising renewable source of cells for therapeutic applications. However, the practical application for neuronal cell replacement is limited by heterogeneity of NSC progeny, relatively low yield of neurons, predominance of astrocytes, poor survival of donor cells following transplantation and the potential for uncontrolled proliferation of precursor cells. To address these impediments, we have developed a method for the generation of highly enriched immature neurons from murine NSC progeny. Adaptation of the standard differentiation procedure in concert with flow cytometry selection, using scattered light and positive fluorescent light selection based on cell surface antibody binding, provided a near pure (97%) immature neuron population. Using the purified neurons, we screened a panel of growth factors and found that bone morphogenetic protein-4 (BMP-4) demonstrated a strong survival effect on the cells in vitro, and enhanced their functional maturity. This effect was maintained following transplantation into the adult mouse striatum where we observed a 2-fold increase in the survival of the implanted cells and a 3-fold increase in NeuN expression. Additionally, based on the neural-colony forming cell assay (N-CFCA), we noted a 64 fold reduction of the bona fide NSC frequency in neuronal cell population and that implanted donor cells showed no signs of excessive or uncontrolled proliferation. The ability to provide defined neural cell populations from renewable sources such as NSC may find application for cell replacement therapies in the central nervous system.


Cell Reports | 2015

Differential Connexin Function Enhances Self-Renewal in Glioblastoma

Masahiro Hitomi; Loic P. Deleyrolle; Erin E. Mulkearns-Hubert; Awad Jarrar; Meizhang Li; Maksim Sinyuk; Balint Otvos; Sylvain Brunet; William A. Flavahan; Christopher G. Hubert; Winston Goan; James S. Hale; Alvaro G. Alvarado; Ao Zhang; Mark Rohaus; Muna Oli; Vinata Vedam-Mai; Jeff M. Fortin; Hunter S. Futch; Benjamin Griffith; Qiulian Wu; Chun hong Xia; Xiaohua Gong; Manmeet S. Ahluwalia; Jeremy N. Rich; Brent A. Reynolds; Justin D. Lathia

SUMMARY The coordination of complex tumor processes requires cells to rapidly modify their phenotype and is achieved by direct cell-cell communication through gap junction channels composed of connexins. Previous reports have suggested that gap junctions are tumor suppressive based on connexin43 (Cx43), but this does not take into account differences in connexin-mediated ion selectivity and intercellular communication rate that drive gap junction diversity. We find that glioblastoma cancer stem cells (CSCs) possess functional gap junctions that can be targeted using clinically relevant compounds to reduce self-renewal and tumor growth. Our analysis reveals that CSCs express Cx46, while Cx43 is predominantly expressed in non-CSCs. During differentiation, Cx46 is reduced, while Cx43 is increased, and targeting Cx46 compromises CSC maintenance. The difference between Cx46 and Cx43 is reflected in elevated cell-cell communication and reduced resting membrane potential in CSCs. Our data demonstrate a pro-tumorigenic role for gap junctions that is dependent on connexin expression.

Collaboration


Dive into the Loic P. Deleyrolle'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge