Research team

Expertise

Primary cell culture from mouse and human, including immune cells (dendritic cells, macrophages, T-cells) and stem cells (embryonic stem cells, induced pluripotent stem cells, neural stem cells and mesenchymal stem cells). Multi-colour flow cytometric analyses for cellular phenotyping and T-cell analysis. Multi-colour immunocytochemical analyses of cultured (stem) cell populations. Ex vivo genetic engineering of (stem) cell populations using mRNA, plasmid DNA, transposons and lentiviral vectors. Animal models of neuro-inflammation, including the experimental autoimmune encephalomyelitis (EAE) and the cuprizone (CPZ) mouse models applicable for multiple sclerosis research. Cell grafting in brain and spinal cord of rodents. Generation of bone marrow chimeric mice. In vivo non-invasive bioluminescence imaging (monitoring stem cell survival) and magnetic resonance imaging (monitoring white matter lesions). Multi-colour and quantitative immunohistochemical (-fluorescence) analysis of neuro-inflammation.

FWO Research Sabbatical 2023-2024 (Prof. P. Ponsaerts) 01/02/2024 - 31/01/2025

Abstract

The research team of prof. Peter Ponsaerts at the Laboratory of Experimental Hematology (University of Antwerp, UA) has established during the past 5 years multiple 2D and 3D murine and human iPSC-derived neuro-immune cell culture models (including neurons, astrocytes, macrophages and microglia), with a specific focus on inflammation, viral infection, stroke and Parkinson's Disease. Having established these models, the research team of prof. Peter Ponsaerts has already implemented standard cytokine profiling, immunocytochemistry (ICC) and bulk RNA-Seq analyses for cell type characterization and cellular responses to inflammation, trauma or infection. Nevertheless, there is an unmet need to advance their experimental toolbox to characterize 2D and 3D neuro-immune cell culture models. The envisaged research sabbatical at the "Mass Spectrometry Lab" of the "Analytical Biochemistry and Proteomics Research Unit" from the "Center for Advanced Studies and Technology (CAST)" at the "University G. d'Annunzio" in Chieti-Pescara in Italy aims at two educational (E) and two scientific (S) action points: (E1) To learn and understand the base principles of high sensitivity proteomics and metabolomics (including metallomics and lipidomics) analyses using state-of-the-art mass spectrometry instruments and methods. (E2) To learn and apply MaxQuant, Perseus, Ingenuity Pathways Analysis (IPA) and MetaboAnalyst software for omics data analysis. (S1) To perform a longitudinal proteomics and metabolomics study of human (h)iPSC-derived bi- and tri-partite neurospheroids during development and maturation. (S2) To perform a proteomics and metabolomics study of mature hiPSC-derived bi- and tri-partite neurospheroids following viral infection, hypoxic/hypoglycemic stress (stroke) and Parkinson's Disease (PD) mimicking conditions. By completion of this research sabbatical, prof. Peter Ponsaerts will not only have gained the necessary bioinformatics skills to analyse proteomics data, but also will be able to integrate novel proteomics and metabolomics data with scRNA-Seq, ICC and electrophysiological analyses performed by his research team at UA, within current and future research projects.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

BOF Sabbatical 2024 - P. Ponsaerts. 01/02/2024 - 31/01/2025

Abstract

The research team of prof. Peter Ponsaerts at the Laboratory of Experimental Hematology (University of Antwerp, UA) has established during the past 5 years multiple 2D and 3D murine and human iPSCderived neuro-immune cell culture models (including neurons, astrocytes, macrophages and microglia), with a specific focus on inflammation, viral infection, stroke and Parkinson's Disease. Having established these models, the research team of prof. Peter Ponsaerts has already implemented standard cytokine profiling, immunocytochemistry (ICC) and bulk RNA-Seq analyses for cell type characterization and cellular responses to inflammation, trauma or infection. Nevertheless, there is an unmet need to advance their experimental toolbox to characterize 2D and 3D neuro-immune cell culture models. The envisaged research sabbatical at the "Mass Spectrometry Lab" of the "Analytical Biochemistry and Proteomics Research Unit" from the "Center for Advanced Studies and Technology (CAST)" at the "University G. d'Annunzio" in Chieti-Pescara in Italy aims at two educational (E) and two scientific (S) action points: (E1) To learn and understand the base principles of high sensitivity proteomics and metabolomics (including metallomics and lipidomics) analyses using state-of-the-art mass spectrometry instruments and methods. (E2) To learn and apply MaxQuant, Perseus, Ingenuity Pathways Analysis (IPA) and MetaboAnalyst software for omics data analysis. (S1) To perform a longitudinal proteomics and metabolomics study of human (h)iPSC-derived bi- and tri-partite neurospheroids during development and maturation. (S2) To perform a proteomics and metabolomics study of mature hiPSC-derived bi- and tri-partite neurospheroids following viral infection, hypoxic/hypoglycemic stress (stroke) and Parkinson's Disease (PD) mimicking conditions. By completion of this research sabbatical, prof. Peter Ponsaerts will not only have gained the necessary bioinformatics skills to analyse proteomics data, but also will be able to integrate novel proteomics and metabolomics data with scRNA-Seq, ICC and electrophysiological analyses performed by his research team at UA, within current and future research projects.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Functional and molecular profiling of immune-competent neurospheroids under stroke-like conditions. 01/11/2023 - 31/10/2024

Abstract

Due to the enormous impact of stroke on the patient's quality of life and on society, decades of research resulted in the identification of thousands of candidate neuroprotective drugs. Unfortunately, none have led to an effective therapy to date. This can partially be attributed to the lack of in vitro systems able to accurately recapitulate human ischemic responses. Fortunately, the advent of induced pluripotent stem cell (iPSC)-technology has provided novel tools for generating human-based in vitro brain models, namely neurospheroids. Extending the host laboratories' preceding research efforts to generate bi-partite (neurons + astrocytes) and tri-partite (neurons + astrocytes + microglia) human iPSC-derived neurospheroids, I hypothesize that subjection of mature tri-partite neurospheroids to stroke-like conditions (oxygen/glucose-deprivation), in combination with advanced single cell analysis tools and measurement of electrophysiological network activity, will aid to unravel biologically relevant cellular and molecular events in the context of stroke pathology. In this way, the combined cellular and molecular toolbox for neurospheroid culture, manipulation and analysis will in short term pave the way for novel fundamental studies unravelling new pathways and/or potential targets for neuroprotection or repair, and in long-term novel therapeutic approaches for patients with cerebral ischemia.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Towards an Off-the-Shelf Therapy: Tolerogenic Dendritic Cell-derived Extracellular Vesicles for the Treatment of Multiple Sclerosis. 01/12/2022 - 30/11/2024

Abstract

MS is a chronic auto-immune disorder of the central nervous system (CNS) and the leading cause of non-traumatic disabling disease in young adults. Although the exact cause of MS remains to be elucidated, it is currently accepted that both genetic and environmental factors affect complex immunological responses, both in the periphery and the CNS. To date, there is still no cure for MS, but several immune-modifying and/or -suppressive treatments, especially targeting the peripheral immune system, have been developed over time. However, these have varying efficacy, have limited long-term effectiveness, and sometimes life-threatening side effects, thereby underscoring the urgent need for novel therapeutic approaches to be developed and evaluated. Tolerogenic dendritic cells (tolDCs) are professional antigen-presenting cells with immunosuppressive properties, priming the immune system into a tolerogenic or unresponsive state against various (self)antigens. TolDCs are essential in maintenance of central and peripheral tolerance through induction of T cell clonal deletion, T cell anergy, generation and activation of regulatory T-cells (Tregs), as well as the direct modulation of pro-inflammatory environments. For that reason, tolDCs show considerable promise as candidates for specific cellular therapy for treatment of allergic diseases, autoimmune diseases or transplant rejections. In this context, the Laboratory of Experimental Hematology (LEH, Cools' team) recently recruited nine patients for a phase I clinical trial evaluating the potential safety and feasibility of tolDCs for the treatment of multiple sclerosis (MS) (NCT02618902). While no serious adverse events are observed in all treated patients, much remains to be understood about the exact molecular and cellular interactions these therapeutic cells provoke in vivo. Accumulating evidence shows that extracellular vesicles (EVs) secreted by immune cells play a key role in intercellular communication. In this project, we aim to determine the immune-regulatory mechanism by tolDCs, hypothesising that it would be mediated by EVs and their immunosuppressive cargo. To achieve this goal, we will apply an unbiased multi-omics approach, both in vitro and in vivo, to unravel the therapeutic potential of tolDC-derived EVs. We hereby anticipate that our findings will lead to ground-breaking insights on current understanding of EVs in immune-regulatory therapy and ultimately will lead to the development of a novel (non-cellular) off-the-shelf therapeutic compound to be evaluated in patients suffering from detrimental auto-immune disorders, including MS. This highly innovative application addresses the use of tolDC-derived EVs as disease-modifying treatment in MS and is expected to provide new insights into how immune tolerance is initiated following interaction of key immune-regulatory cargo of the EVs with peripheral and CNS immune cells. With this project, we present a clinically relevant project relying on inventive fundamental research with a high translational value and valorization potential. The integrative multiomics analysis will give a better insight in the molecular pathways involved in the induction of tolerance and immunoregulation by tolDCs. Furthermore, this project could lead to the development of a cell-free therapy based on tolDC-EV for the treatment of MS, which can surpass drawbacks associated with cell therapy. In addition, the possibility of allogenic exosome therapy would result in a more positive cost-benefit ratio since an "off-the-shelf" product is less expensive than an individualized cell therapy. Hence, the study proposed here is merely the beginning of numerous possible new research questions as well as clinical translation.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

SECRET - Evaluation of the neuro-protective properties of perinatal derivatives using murine iPSC-derived neurospheroids. 01/12/2022 - 30/11/2024

Abstract

Perinatal tissues encompass a large and diverse cell/tissue family obtained from term pregnancies. Perinatal derivatives (PnDs) include amniotic membrane, chorion, Wharton's jelly, amniotic fluid, and the cells isolated from these tissues, as well as the factors that these cells release. PnD have drawn much attention due to their immune-modulatory and tissue-protecting properties, making them attractive candidates in regenerative medicine. Focussing on neuro-protection, we will here investigate whether different types of PnDs are able to inferfere with inflammation-associated neuro-degenerative processes in murine induced pluripotent stem cell (iPSC)-derived neurospheroids. Neurospheroids cultured from iPSC represent an important research tool to study neuron-astrocyte interactions, during development, homeostasis and stress. Hereto, we developed a 5-week old murine iPSC-derived neurospheroid model containing mature neurons and astrocytes in order to evaluate the therapeutic potential of several neuro-protective/modulating compounds in vitro preceding animal experiments. Following development and characterization of this new murine iPSC-derived neurospheroid model, its sensitivity to immune signal-induced stress (a.o. stimulation with IL1b, TNF and/or LPS) has been demonstrated by monitoring astrocyte activation (a.o. production of IL6 and CXCL10). In this new project in collaboration with the 'Universita Cattolica del Sacre Cuore' (Rome, Prof. Ornella Parolini) and the 'Centro di Ricerca E. Menni' (Brescia, Dr. Antonietta Silini), we will now investigate the neuro-protective/modulating activity different PnDs on astrocyte activation in murine iPSC-derived neurospheroids. These studies will then allow to preselect the most potential PnDs for subsequent animal studies and/or human clinical trials in the field of neuro-degenerative disease.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Towards an end-to-end solution for unbiased cellular phenotyping of intact cerebral organoids. 01/11/2022 - 31/10/2024

Abstract

The cerebral organoid is an emerging model system with high potential for both fundamental and applied neuroscience research. However, batch-to-batch variability and the inability to characterize these specimens at the cellular level with high throughput, hampers their integration in an industrial setting. With this project, we intend to develop a pipeline that enables unbiased cellular phenotyping of intact cerebral organoids by using a combination of multiplex fluorescent labelling, light-sheet microscopy, and deep learning. Our approach builds on the concept that cells can be accurately identified by means of sheer morphological information. First, we will perfection cell profiling in co-cultures of different brain cell lines by training machine learning-based classifiers. Then, we will translate the concept to 3D, using spheroids from the same cells. To this end, we will render the staining compatible with chemical clearing and conceive a sample mounting procedure for serial, isotropic image acquisition. Finally, we will deploy the optimized method to recognize cell type and state in iPSC-derived cerebral organoids that have been challenged with selected compounds or seeded with glioblastoma cells. The approach will add to a more standardized quality-control of organoids and will facilitate the adoption of this model in drug-screening pipelines. Ultimately, this will boost the translatability of preclinical research and lower attrition rates in late-stage clinical trials.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Comprehensive phenotyping of neuro-organoids by deep learning. 01/11/2022 - 31/10/2024

Abstract

Identification of disease mechanisms and novel therapeutic targets relies on the use of cell culture and animal models. While the former are overly simplified, the latter are not human and ethically contested. Suboptimal models at the discovery side will inevitably lead to a steep loss of leads in clinical trials. With the advent of human induced pluripotent stem cell technology, it has now become possible to generate organoids that more faithfully capture part of the heterogeneity and three-dimensional context of human tissue. Several research labs at the University of Antwerp (UA) recognize their potential and have therefore implemented a variety of human patient-derived organoid cultures, in particular for neuroscientific research lines. However, batch-to-batch variability and the inability to characterize these specimens at the cellular level with high-throughput, hamper their integration in a routine screening setting. Therefore, we have the ambition to develop an end-to-end solution that enables unbiased cellular phenotyping of intact neuro-organoids by using a combination of fluorescent labelling, advanced microscopy, and artificial intelligence (AI).

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Optimization and pharmacokinetics of allele-specific antisense oligonucleotide therapy for late-onset sensorineural hearing impairment DFNA9. 01/10/2022 - 30/09/2025

Abstract

Hearing loss (HL) is a growing problem in modern society, and is associated with an increased risk for social isolation and unemployment. Although the genetic basis of adult-onset HL is still largely elusive, heritability is suggested in 30-70% of cases. Lacking curative or preventive (genetic) treatments, hearing aids and cochlear implantation (CI) can relieve part of the burden of HL. However, the majority of patients with adult-onset HL still do not experience a satisfactory improvement of their auditory function with these devices. Furthermore, the outcome of CI in adult-onset cases is often less favorable as compared to CI in congenital HL cases. DFNA9, caused by mutations in the COCH gene, is amongst the best-studied forms of dominantly-inherited adult-onset HL. The c.151C>T (p.(P51S)) mutation likely occurred many generations ago, and is now estimated to cause adult-onset progressive HL and vestibular dysfunction in >1500 Dutch and Belgian individuals. The high prevalence of this founder mutation in our cohorts presents a unique opportunity to overcome the translational obstacles in the development of novel inner ear therapeutics. The adult onset of hearing loss provides a window of opportunity for therapeutic intervention. The large cohort of patients with the exact same mutation provide enough power for future clinical trials. The dominant inheritance pattern of DFNA9 implies that only one of the two gene copies (alleles) contains a mutation. These DFNA9 mutations in the COCH gene are all well-established to result in the production of toxic cochlin proteins that interfere with the function of the healthy cochlin proteins produced from the healthy allele. As such, a treatment that can block the formation of these toxic cochlin proteins has high therapeutic potential, especially when administered in an early stage of the disease. The remaining cochlin proteins produced from the healthy allele are sufficient for normal inner ear function. Recently published antisense oligonucleotides (AONs; small strands of synthetic DNA and RNA molecules) can specifically induce the degradation of c.151C>T mutant COCH transcripts, but not COCH transcripts resulting from the healthy allele (de Vrieze et al, Molecular Therapy – Nucleic Acids, 2021). In this project, we aim to further improve the efficiency and stability of our best-performing c.151C>T AON by introducing chemical modifications, and perform a series of pre-clinical validation studies in patient-derived stem cell models and a humanized DFNA9 mouse model. These data will provide a strong foundation for a swift translation of our AON treatment to future clinical trials. As there is virtually no prior art on the use of AONs to treat inner ear disorders, our studies are designed to also provide insights in the safety and feasibility of AONs a treatment paradigm of inner ear disorders in general.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Refining the efficacy of systemic administration of bioactive molecules for Parkinson's Disease (RePark) 01/09/2022 - 31/08/2025

Abstract

Parkinson's Disease (PD) is a neurodegenerative disease primarily linked to ageing affecting the psychomotor functions. An impairment in the activity of dopaminergic neurons (aggregation and intracellular accumulation of alfa-synuclein), is at the basis of this functional loss. Current therapies do not revert the PD progression but stand on the relief of the symptoms. Importantly, the efficacy of new therapeutic approaches is hampered by the lack of in vitro models that mimic the PD's hallmarks. Moreover, the effectiveness of therapeutic approaches is hampered by the low ability of the drugs to cross the blood- brain barrier (BBB). RePark will develop an in vitro PD model that combines a mimic of the BBB, the brain's extracellular matrix (ECM) and brain cells. With this system it will be possible to overcome the disadvantages of using animal models and to recapitulate in vitro the cellular characteristics of PD, as well as to assess the delivery and efficacy of new therapeutic drugs.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Bioreactor infrastructure for upscaled culture of organoids and tumoroids. 01/06/2022 - 31/05/2024

Abstract

In this application, we request financing for three benchtop CERO 3D Cell Culture Bioreactor units for the culture of 3D cell cultures, including spheroids and organoids, that are increasingly being used in biomedical research. Currently, 3D organoids and spheroids are cultured in traditional cell culture plates under static or shaking (using orbital shaker) conditions in a standard CO2 cell culture incubator, which is suboptimal for long-term and large-scale culture of spheroids and organoids. A bioreactor system would take organoid and spheroid culture at the campus to a next level in terms of quality (improved viability, maturation and homogeneity) as well as quantity. Each CERO 3D Cell culture bioreactor unit can maintain four 50 mL organoid cultures, including monitoring and control of temperature, pH and carbon dioxide levels. In total, the envisaged bioreactor infrastructure will be able to accommodate twelve simultaneous organoid cultures under highly controlled conditions. The envisaged CERO 3D Bioreactor units will be applied for multiple research domains at the University of Antwerp, and more specifically for upscaled culture of stem cell-derived spheroids and organoids, tumoroids derived from primary tumour material of patients, stem cell-derived cardiomyocytes, stem cell-derived cartilage tissue and intestinal organoids. Furthermore, based on our own experience in upscaled organoid culture, the instalment of bioreactor units has become an urgent need to progress towards future valorisation activities.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Flow Cytometry and Cell Sorting Core Facility Uantwerpen (FACSUA) 01/01/2022 - 31/12/2026

Abstract

Flow cytometry is a widely used technique that allows the simultaneous and multi-parameter analysis of physical and biochemical characteristics of a population of living cells or particles in a heterogenous sample. The Laboratory of Experimental Hematology (LEH) has 20+ years of demonstrable experience in flow cytometry (200+ published manuscripts), as well as experience in guidance and support of both internal and external research groups with flow cytometric experiments (30+ joint manuscripts). With this application, we now aim to maintain and expand a flow cytometry and cell sorting core facility at the AUHA. The ambition of LEH is to make basic and advanced flow cytometry available for all active and prospective users at the AUHA in order: (i) to leverage qualitative cell biological and (pre)clinical cellular research, (ii) to provide qualitative education covering flow cytometry and its applications over multiple faculties (FGGW, FBD and FWET), and (iii) to provide external service using flow cytometry as a basis, both intellectually as well as practically.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Tau-induced senescence in human mini-brain. 01/10/2021 - 30/09/2025

Abstract

Defects in the microtubule associated protein tau typify a range of neurodegenerative disorders termed tauopathies, which includes Alzheimer's disease. Recent studies point to the potential involvement of cellular senescence, an irreversible non-proliferative state, associated with inflammatory cytokine secretion, in disease development. However, the causality, timing, and afflicted cell types remain poorly characterized. The goal of this project is to define the exact causal relationship between senescence and tauopathy development in a human context. To achieve this, I intend to produce human iPSC-derived brain organoids that contain the three major cell types of the brain (neurons, astrocytes, or microglia) and assess the emergence of senescence therein using deep coverage microscopy and single cell sequencing. Once a cell-specific senescence signature has been established, I will measure its penetrance in mutant organoids that recapitulate the hallmarks of tau pathology. Finally, I will evaluate whether senescence-targeting compounds modulate tau pathology and influence organoid condition. Together, this work should allow defining whether senescence is a driving factor in human tau pathology development and unveil its potential as druggable node.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Immunological control of Varicella zoster virus (VZV)-infected iPSC-derived brain models by steady-state and immune-compromised astrocytes and microglia. 01/01/2021 - 31/12/2024

Abstract

Varicella zoster virus (VZV) is a member of the herpesvirus family and is a highly successful and ubiquitous human pathogen. Both in children and in adults, varicella-related complications may lead to hospitalisation. While in children direct neurological complications may occur following primary infection (varicella), in adults vasculitis and neurological complications are not uncommon following reactivation of latent VZV (herpes zoster). With a clear link between VZV and neuropathology, it is inevitable that the immune system of the central nervous system (CNS) will be challenged by VZV. However, to date little is known about how astrocytes and microglia behave upon encounter of VZV in the CNS. In this project, we will address this question using an established human in vitro model of axonal infection of human induced pluripotent stem cell (hiPSC)-derived CNS neurons with fluorescent reporter VZV stains. Using this model, we will first longitudinal monitor how hiPSC-derived astrocytes and microglia influence the processes of VZV infection, latency and reactivation. Next, using iPSC models derived from VZV patients with mutations in POLRIII, we will investigate whether immune compromised astrocytes and/or microglia can control neuronal VZV infection. Altogether, these studies will help us understanding innate immune control of VZV in the CNS, and will allow - beyond the scope of this project – to develop novel strategies to prevent VZV spreading in the CNS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Deep phenotyping of cellular heterogeneity and maturation in human iPSC-derived brain organoids and cardiomyocytes. 01/01/2021 - 31/12/2024

Abstract

Pluripotent stem cell (PSC) technology is increasingly gaining interest for modelling diseases and developing precision therapeutics. However, the immaturity and heterogeneity of PSC-derived cell populations impinge on the reproducibility and preclude their use for comparative and diagnostic analyses. Recognizing these caveats for their own human induced (hi)PSC-based research, the Laboratories of Experimental Hematology, Cardiogenetics and Cell Biology and Histology have teamed up to develop a pipeline that enables accurate phenotypic staging of hiPSC-derived cell culture models. Specifically, to create mature and standardized 2D and 3D-models for cardiomyocyte cultures and brain organoids, our consortium proposes to optimize the culture conditions and refine the interrogation methods. Longitudinal follow-up and validation of functional activity in the differentiation products will be achieved by means of high-throughput multi-electrode array recordings and live cell calcium and voltage imaging. In parallel, cellular heterogeneity will be mapped with quantitative immunofluorescence and transcriptome analyses. The correlation of functional and molecular readouts will allow establishing a biomarker panel for mature hiPSC-derived cardiomyocyte models and brain organoids. Thus, with this work, we intend to develop more reproducible models and to allow selecting only those with the highest functional maturity, a prerequisite for our future stem cell research aimed at studying and treating neuro-inflammatory and cardiogenetic disorders.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Upgrade of 9.4T Bruker BioSpec MRI imaging system to Avance NEO hardware architecture. 01/05/2020 - 30/04/2024

Abstract

Upgrade of the hardware of existing equipment (9.4T MRI system from Bruker) to perform state of the art MRI investigations in the brain of small animals such as mice, rats and birds. This hardware upgrade will enable implementation of all new Bruker software packages.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Multi-well microelectrode array (MEA): a bridge to highthroughput electrophysiology. 01/05/2020 - 30/04/2024

Abstract

This project aims to upgrade the current electrophysiology technologies at UAntwerpen by acquiring a state-of-the-art MicroElectrode Array platform (MEA). To study the electrophysiological properties of excitable cells, currently patch-clamping is the gold standard. However, this is an extremely labour-intensive and invasive technique, limited to short-term measurements of individual cells at single time points. On the other hand, MEAs enable high-throughput non-invasive longitudinal real‐time measurements of functional cellular networks, without disrupting important cell-cell contacts, and thus provide a more physiologically relevant model. The multi-well format allows repeated recordings from cell cultures grown under various experimental conditions, including the opportunity to rapidly screen large drug libraries. Based on these advantages, multi-well MEAs are the most suitable instrument for functionally elucidating the pathomechanisms of neurological/cardiac disorders by performing (1) cardiac activity assays: measurement of field and action potentials from (iPSC-)cardiomyocytes to investigate wave-form, propagation and irregular beating; (2) neural activity assay based on three key measures: frequency of action potential firing, synchrony as measure for synaptic strength and oscillation as hallmark for neuronal organization in time; (3) (iPSC-)vascular smooth muscle contractility assay based on impedance alterations.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Modulation of astrocyte activation in murine iPSC-derived neurospheroids. 01/01/2023 - 31/12/2023

Abstract

Neurospheroids cultured from induced pluripotent stem cells (iPSC) represent an important research tool to study neuron-astrocyte interactions, during development, homeostasis and stress. In course of the EU MSCA ITN PMSMatTrain project, the UAntwerp partner developed a 5-week old murine iPSC-derived neurospheroid model containing mature neurons and astrocytes in order to evaluate the therapeutic potential of several neuro-protective/modulating compounds in vitro preceding animal experiments. Following development and characterization of this new murine iPSC-derived neurospheroid model, its sensitivity to immune signal-induced stress (a.o. stimulation with IL1b, TNF and/or LPS) has been demonstrated by monitoring astrocyte activation (a.o. production of IL6 and CXCL10). This SEP grant will be applied to support the 4th PhD year of Julia Di Stefano in which the candidate will investigate the neuro-protective/modulating activity of APRIL (delivered by means of AAV vectors) and placenta stem cell-derived factors (delivered by means of extracellular vesicles) on astrocyte activation in murine iPSC-derived neurospheroids. Finally, it is expected that the technology developed here will become an important additional tool in (i) fundamental research to study neuro-development and functioning, as well as (ii) to preselect interesting therapeutic molecules before proceeding to animal studies and/or human clinical trials.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Development and validation of a bona fide iPSC derived human neuronal infection model to evaluate antivirals targeted against neurotropic viral infections. 01/05/2022 - 30/04/2023

Abstract

Neurotropic viral infections continue to cause major disease and economic burden. Such infections are most commonly caused by herpesviruses, arboviruses and enteroviruses, often leading to severe neurological damage with poor clinical outcomes. The search for interventions to prevent and/or treat these infections is however challenging. The main reason for this is the nature of the target cells, neurons, which are chiefly non-renewable and drastically differ from other cells (or cell lines). Discovery of novel antivirals via the classically performed research with cell lines, is not appropriate for viruses that infect neurons. Highly specialized, bona fide, human neuronal culture models are imperative. With this project, we will develop specialized neuronal cultures aimed at higher throughput antiviral screening.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Development of allele-specific CRISPR-nuclease gene therapy for late-onset sensorineural hearing impairment in a new humanized DFNA9 mouse model. 01/01/2020 - 31/12/2020

Abstract

Hearing impairment is the most frequent sensory deficit in the human population, affecting 440 million people worldwide, whereby loss of hearing and balance has a significant impact on quality of life and society. Hearing loss is also listed by the World Health Organization as a priority disease for research into therapeutic interventions to address public health needs. DFNA9 (DeaFNess Autosomal 9) is an autosomal dominant hearing disorder caused by a heterozygous gain-of-function mutation in the COCH gene (Coagulation Factor C Homology) and is characterized by progressive late-onset (3rd-5th decade) sensorineural hearing loss (SNHL) and deafness. At current, it is believed that the presence of aberrant COCH proteins in the extracellular matrix (ECM) of the inner ear leads to local cell damage resulting in progressive hearing loss. Within Belgium and the Netherlands, there are > 1000 patients affected by the P51S COCH mutation, who – in the current absence of a disease modifying therapy – will develop deafness and balance loss. Furthermore, there are over twenty different COCH mutations identified in people from all over the world that lead to SNHL. Given the genetic nature of this disorder with highly specific mutations, as well as recent advances in CRISPR-nuclease mediated gene therapeutic approaches, there is a great opportunity to develop a successful therapeutic strategy to reduce or prevent DFNA9-induced SNHL.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Novel Biomaterial-based Device for the Treatment of Progressive MS - An Integrated Pan- European Approach (PMSMatTrain). 01/05/2019 - 31/01/2024

Abstract

PMSMatTrain is focusing on gaining a comprehensive understanding of the progressive (late degenerative phase) of multiple sclerosis (PMS) from basics to translation, fully supported by 8 beneficiaries (6 research institutions, 2 SMEs). Recruited ESRs will receive compulsory discipline-specific, generic and complementary transferable skills training. PMSMatTrain's Joint Research Education and Training programme (JRTP) will provide early stage researchers with high quality research and transferable skills training in intellectual property, leadership skills, innovation, regulatory affairs, entrepreneurship, gender policy, and medical device evaluation, which will ensure that they are immediately employable in industry. The consortium will develop a multi-modal hyaluronan-based medical device designed to release small molecular weight anti-inflammatory molecules (APRIL and sPIF) followed by remyelination and neuroprotective drugs (ibudilast and miconazole). PMSMatTrain will for the first time utilise these functionalised multi-modal biomimetic hyaluronan scaffolds as a tool to investigate cross-talk between signals arising due to chronic neuroinflammation and those leading to demyelination and axonal loss, while identifying molecular mechanisms that facilitate remyelination and neuroprotection in PMS. This approach could yield the first cortex-proximal and directed biomaterials-based disease-modifying therapy for PMS. These scaffolds will be tested in state of the art MS patient induced stem cell-derived oligodendrocyte cultures and organotypic cultures to investigate MS pathophysiology. In vivo responses will be characterised using field-leading MRI and mass spectrophotometry protocols. PMSMatTrain will also generate a clinically-relevant in silico model of drug elusion and dispersal within the CNS. Our industry partners will develop the end-device by providing standardised manufacturing protocols for scaled-up production and commercialisation of the cGMP product.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

A human in vitro demyelination model for studying the effect of M2 macrophages/microglia. 01/01/2019 - 31/12/2020

Abstract

The host's laboratory's promising results of IL13 treatment in a mouse model for MS, encourage us to further investigate this potential therapeutic strategy in a human setup. Therefore, we propose to develop a human in vitro demyelination model to study the effect of IL13-induced M2 macrophages and microglia. We will do so by (I) differentiating hiPSC towards oligodendrocytes; (II) evaluating the myelinating potential by oligodendrocyte-neuron co-culture; (III) optimizing a toxin-induced demyelination procedure; and (IV) evaluating the effect of macrophage/microglia addition with or without IL13 stimulation.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Inner ear gene therapy to prevent deafness in DFNA9. 01/10/2018 - 30/09/2022

Abstract

Hearing loss has a significant impact on quality of life and society in general. Hearing impairment is the most frequent sensory deficit in human populations, affecting 360 million people worldwide. It was listed by the World Health Organization as one of the priority diseases for research into therapeutic interventions to address public health needs. DFNA9 is a dominant hereditary disorder, caused by heterozygote mutations in the COCH gene, which progressively leads to bilateral deafness and balance loss by the age of 50-70 years. Currently, no treatment is available to prevent hearing loss or balance loss in DFNA9 patients. Local gene therapy to restore hearing or prevent hearing loss has been studied in neonatal mouse models for several years. Currently, a clinical study is ongoing in adult patients with profound hearing loss to restore hair cells by injecting virus-based vectors -carrying correcting genetic information- directly into the inner ear. In this project, we aim to generate an inner ear gene therapy tool to prevent hearing loss in a pre-clinical mouse model of DNFA9. Using Adeno-associated virus (AAV)-based vectors, we will apply CRISPR-Cpf1 genome engineering technology to target directly within in the cochlea Coch genomic DNA in a safe and effective way in order to disrupt expression of the mutant (and wild type) Coch protein before onset of disease. Hereby, we expect to reduce or prevent DNFA9-associated sensorineural hearing loss.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Development of next-generation 3D brain organoids for the study and modulation of immunemediated neurodegeneration in cerebrovascular disease. 01/01/2018 - 31/12/2022

Abstract

Developing novel neuroprotective and/or immune-modulating therapeutic strategies for almost every neurological disease or trauma requires, both for academia and pharmaceutical industry, the existence of robust in vitro cell culture models to mimic disease-associated pathological events. Unfortunately, a complex interplay between multiple central nervous system (CNS) cell types and multiple cell types from the body's peripheral immune system, cannot be easily recapitulated by currently used 2-dimensional (2D) co-culture assays. It is exactly therefore that successful pre-clinical experimental efficacy has proved to be very difficult to translate into clinical benefit, and as a consequence there is an increasing gap in knowledge and progress between bench and bed side. One highly promising novel approach to improve the predictive power of in vitro human neuro-immune research consists in developing modular 3D brain organoids that resemble brain tissue at the structural, cellular and functional level. Within this project we aim to develop and optimize a new method for generating isogenic 3D brain organoids, comprising human pluripotent stem cell (hPSC)-derived neurons, astrocytes and microglia. Furthermore, hPSC-derived astrocytes and endothelial cells will be used to create a blood-brain-barrier model for physical separation of hPSC-derived macrophages from the generated human 3D brain organoids. Together, this integrated cell system will represent a powerful new 3D human neuro-immune cell culture paradigm. Within this multidisciplinary IOF-SBO project, the methodological approach to generate 3D brain organoids, combined with the experience in the field of clinical research and the availability of patient samples, is truly unique and will - in first instance - highly contribute to the field of in vitro cerebrovascular disease modelling and treatment validation. Furthermore, our aims to install an integrated 3D brain organoid technology platform at the University of Antwerp, will - given the current scientific and economic interests – allow for both short-term and long-term valorisation of our combined efforts, with both intellectual (PhD-theses, A1 publications) as well as financial (contract research) revenues.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Human and murine macrophages with a stable alternative activation as a therapeutic tool to promote neuroregeneration. 01/01/2018 - 31/12/2021

Abstract

Worldwide, spinal cord injuries (SCI) are a major cause of morbidity, mortality and reduced quality of life. Until now no regenerative therapy for SCI is available and standard care of SCI patients includes primarily the administration of immunosuppressant drugs and rehabilitation. Thus, new therapeutic strategies are desperately needed. After SCI, pro-inflammatory macrophages dominate the spinal cord and exert detrimental effects by secreting multiple pro-inflammatory factors, by stimulating the formation of the inhibitory fibrotic scar, and by attacking dystrophic axons. Previously, the applicants demonstrated that the implantation of IL-13-expressing mesenchymal stem cells or macrophages induces 'antiinflammatory' arginase-1 (Arg-1)-positive macrophages/microglia in the injured spinal cord, leading to improved functional recovery. Therefore, we investigate here whether high Arg-1 production by macrophages with a stable alternative activation leads to the suppression of pro-inflammatory macrophages/microglia after SCI via arginine depletion. In addition, for translation to the human situation, we will investigate whether Arg1-overexpressing human macrophages exert neuroprotective effects in an in vitro multicellular co-culture model of human stem cell-derived corticospinal-like motor neurons, astrocytes and microglia. Thus, in this project, we aim to analyse one key mechanism of how anti-inflammatory macrophages improve functional recovery after SCI.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Application of human 3D brain organoids to evaluate the potency of interleukin 13 for modulation of detrimental microglia and macrophage immune response. 06/12/2017 - 31/12/2019

Abstract

Due to the current understanding that multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS), where mononuclear cell infiltration in brain and spinal cord is a major contributor to demyelination, gliosis, axonal loss and eventually loss of neuronal function, we investigated over the past 6 years whether local modulation of CNS lesions with the immune-modulating cytokine interleukin (IL)13 might ameliorate detrimental disease progression. While our pre-clinical studies in the cuprizone (CPZ)-induced CNS inflammation/demyelination mouse model for human MS have demonstrated proof-of-principle for this approach, currently we do not know whether human microglia and macrophages are equally well susceptible to IL13-mediated immunomodulation in the pro-inflammatory MS environment. Using advanced human induced pluripotent stem cell (hiPSC) derived 3D cell culture models, we aim to provide further pre-clinical rationale for the use of IL13 as an additional treatment approach in advanced stage MS.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Gene Therapy for DFNA9 : downregulating the mutant COCH gene in mammalian cell lines by uising a synthetic adeno-associated viral vector Anc80L65 and CRISPR/Cas9-mediated genetic editing. 01/11/2017 - 31/10/2018

Abstract

DNFA9 is a cause of autosomal dominant (AD) non-syndromic late-onset sensorineural hearing loss (SNHL) associated with progressive bilateral vestibular failure (BVF). The age of SNHL onset varies depending on the mutation though the average onset age lies around 3rd-5th decade. It typically starts as downsloping of the audiogram at the age of onset and evolution towards deafness. DFNA9 is caused by mutations in the COCH gene (Coagulation Factor C Homology), which is located on chromosome 14q12-13 and encodes for a 550 amino acid protein, cochlin, which is expressed throughout the inner ear in spindle-shaped cells located along nerve fibers between the spiral ganglion and sensory epithelium. Over twenty mutations have been identified in regions, including North America, Japan, Australia, Korea, China and Belgium/Netherlands. Our objective is to establish an in vitro proof-of-principle for a gene therapeutic approach that targets mutant cochlin expression in the inner ear using Anc80L65AAV/CRISPR/Cas9-mediated gene editing. We hope to establish in vitro that this technique enables specific correction or downregulation of the mutant COCH gene in mammalian cell lines without modulating the normal COCH allele, which is still present in this heterozygous disorder. This work can provide proof-of-concept for in vivo studies in transgenic heterozygous COCH mice targeting the mutated COCH gene by means of an AAV.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Development of isogenic human embryonic stem cell-derived 3D neuro-immune cell culture models: pre-clinical evaluation of interleukin 13 for microglia and macrophage immunomodulation under stroke-like pathology. 01/10/2017 - 30/09/2021

Abstract

Development of three-dimensional (3D) in vitro cell culture models for human neuro-immunological research is currently a hot topic in medical cell biology research. Although multiple protocols have been described for generating human 3D brain organoids starting from pluripotent stem cells, current models display several limitations, including the lack of extracellular matrix (ECM), the absence of multiple types of immune cells and a functional blood-brain-barrier (BBB). With this project we aim to develop and optimize a new method for generating 3D neuro-immune cell culture models to study and modulate human neuro-inflammatory responses. For this, isogenic 3D cell cultures comprising human embryonic stem cell (hESC)-derived neurons, astrocytes and microglia will be established on decellularized mouse brain sections in order to provide growth and organizational support by original brain ECM proteins. In addition, hESC-derived astrocytes and endothelial cells will be used to create a BBB model for physical separation of hESC-derived macrophages. Further inclusion of genetic engineering strategies, to allow for real time bioluminescence imaging and (live cell) confocal microscopy, will be applied to ensure profound validation and high throughput screening applications. Once established, we will use this technology to further extend our research efforts to optimize therapeutic strategies based on interleukin (IL)13-mediated immunomodulation, following hypoxic and hypoglycemic stress (i.e. stroke-like conditions). Once validated, we believe that implementation of the proposed 3D brain organoid technology by academia and/or pharmaceutical industry will not only have great impact on the reliability of pre-clinical drug screening, and consequently on the medical and social investments associated with patient care, but also will find application in advanced human toxicology research.

Researcher(s)

Research team(s)

Project website

Project type(s)

  • Research Project

Development of isogenic human embryonic stem cell-derived 3D neuro-immune cell culture models: pre-clinical evaluation of microglia and macrophage immunomodulation for stroke treatment. 01/10/2017 - 30/09/2020

Abstract

Development of three-dimensional (3D) in vitro cell culture models for human neuro-immunological research is currently a hot topic in medical cell biology research. Although multiple protocols have been described for generating human 3D brain organoids starting from pluripotent stem cells, current models display several limitations, including the lack of extracellular matrix (ECM), the absence of multiple types of immune cells and a functional blood-brain-barrier (BBB). With this project we aim to develop and optimize a new method for generating 3D neuro-immune cell culture models to study and modulate human neuro-inflammatory responses. For this, isogenic 3D cell cultures comprising human embryonic stem cell (hESC)-derived neurons, astrocytes and microglia will be established on decellularized mouse brain sections in order to provide growth and organizational support by original brain ECM proteins. In addition, hESC-derived astrocytes and endothelial cells will be used to create a BBB model for physical separation of hESC-derived macrophages. Further inclusion of genetic engineering strategies, to allow for real time bioluminescence imaging and (live cell) confocal microscopy, will be applied to ensure profound validation and high throughput screening applications. Once established, we will use this technology to further extend our research efforts to optimize therapeutic strategies based on interleukin (IL)13-mediated immunomodulation for cerebrovascular disease.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Generation of iPSC-derived 3D neuro-immune cell culture models for study and modulation of neuro-inflammatory processes. 01/01/2017 - 31/12/2020

Abstract

Development of three-dimensional (3D) in vitro cell culture models for neuroscience research is currently a hot topic in medical cell biology research. Although multiple protocols have been described for generating 3D brain organoids starting from pluripotent stem cells, current models display several limitations, including the lack of extracellular matrix (ECM) and the absence of glial and immune cells. With this project we aim to develop and optimize a new method for generating 3D neuro-immune cell culture models to study and modulate neuro-inflammatory responses. For this, 3D cell cultures comprising induced pluripotent stem cell (iPSC)-derived neurons, astrocytes and microglia will be established on decellularized mouse brain sections in order to provide growth and organizational support by the original brain ECM. Further inclusion of genetic engineering strategies, to allow for real time bioluminescence imaging and live cell confocal microscopy, will be applied to ensure high throughput screening applications. Once established, we will use this technology to further extend our research efforts to optimize therapeutic strategies based on interleukin (IL)13-mediated immunomodulation, following hypoxic and hypoglycemic stress. Once validated, we believe that implementation of this technology by pharmaceutical industry will have great impact on the reliability of pre-clinical drug screening, and consequently on the medical and social investments associated with patient care.

Researcher(s)

Research team(s)

Project type(s)

  • Research Project

Unravelling the role of neuroglobin in neuroprotection and neuroplasticity. 01/10/2016 - 30/09/2018

Abstract

The general objective is to obtain insight in the contribution and working mechanism of two vital processes in the brain: 1) neuroprotection and 2) 17β-estradiol (E2) induced neuroplasticity. As we aim to focus on different expression patterns of Ngb both in vitro and in vivo, the first objective is to improve valid Ngb overexpressing and knock-out (KO) systems.

Researcher(s)

Research team(s)

    Project type(s)

    • Research Project

    Modular confocal microscopy platform with light sheet illumination. 01/05/2016 - 30/04/2020

    Abstract

    The application concerns an innovative microscopy platform for visualizing cells, tissue specimen and living small model organisms in three dimensions at unprecedented speed and with excellent resolution and contrast. As a unique feature, the platform is equipped with a light-sheet module, which is based on an orthogonal configuration of laser-generated, micrometer-thin plane illumination and sensitive one-shot detection. Seamless integration with confocal modalities enables imaging the same sample from the micro- to the mesoscale. The device has a broad application radius in the neurosciences domain inter alia for studying neurodegeneration and -regeneration (e.g. whole brain imaging, optogenetics); but it also has direct utility in various other fields such as cardiovascular research (e.g. plaque formation and stability), plant developmental research (e.g. protein localization during plant growth) and ecotoxicology (e.g. teratogenicity and developmental defects in zebrafish). Furthermore, its modular construction will enable adaptation and targeted expansion for future imaging needs.

    Researcher(s)

    Research team(s)

    Project type(s)

    • Research Project

    Improving the efficacy of peripheral anti-glioma vaccination by local modulation of the tumour's immunosuppressive microclimate. 01/10/2015 - 30/09/2018

    Abstract

    In this doctoral research proposal, we propose to combine both innate and adaptive immune stimulation approaches in order to mount a more potent anti-tumour response. In addition, an in-depth characterisation of central nervous system and peripheral immune changes will be performed following tumour induction and/or anti-tumour vaccination. With this research project we therefore aim to contribute to a better understanding of cellular interactions in the events of tumour growth and eradication, and to the improvement of current immunotherapeutic interventions of GBM.

    Researcher(s)

    Research team(s)

      Project type(s)

      • Research Project

      Unravelling the cellular and molecular determinants of neuroprotection following implantation of interleukin 13-expressing mesenchymal stem cells in inflammatory brain tissue. 01/10/2015 - 31/12/2016

      Abstract

      Detrimental inflammatory responses in the central nervous system (CNS) are a hallmark of various neurodegenerative pathologies, with multiple sclerosis (MS) and stroke/trauma being excellent examples demonstrating the highly complex interplay between CNS resident microglia and lesion infiltrating leucocytes. Lesion-associated inflammatory responses, both in the acute as well as in the chronic phase, are classified as being highly pro-inflammatory. In this context, it is generally believed that a functional conversion of a pro-inflammatory type M1 microglia/macrophage phenotype, which can readily be detected in severe CNS inflammatory lesions, into a type M2a immune modulating microglia/macrophage phenotype can have a beneficial effect on disease outcome. Using our extensive experience with cell implantation into the CNS of mice, our current research aims at in vivo modulation of neuro-inflammatory responses by intracerebral implantation of mesenchymal stem cells (MSCs) genetically engineered to express interleukin (IL)13, a potent inducer of the M2a phenotype in macrophages. Thus far, we have demonstrated in the cuprizone (CPZ) mouse model of CNS inflammation and demyelination that microglial quiescence and subsequent protection against demyelination coincides with the appearance of M2a-polarised macrophages in the CNS following grafting of IL13-expressing MSCs. Continuing our research, this PhD project will focus on unravelling the in vivo signalling events that lead to IL13-mediated induction of the M2a macrophage phenotype within MSC graft-infiltrating macrophages in the CNS, as well as the cellular interactions by which these M2a macrophages can influence the development of microglia/macrophage-mediated neuro-inflammation in the CNS. Upon completion we hope to: (i) further elucidate the immunological consequences of MSC grafting in the CNS, and (ii) provide pre-clinical rationale for the use of IL13 as an immune modulating cytokine in the CNS.

      Researcher(s)

      Research team(s)

        Project type(s)

        • Research Project

        Neuro Image-guided decoding of mechanisms involved in healthy, accelerated and pathological aging. 01/01/2015 - 31/12/2018

        Abstract

        The overall goal of project is to contribute new information that will greatly increase our understanding about underlying mechanisms of hypothalamus-driven normal, accelerated and pathological aging of the brain, with the focus on structural alterations and subsequent alterations of specific functional networks such as the default mode network (DMN) and its anticorrelated networks. Our results will provide fundamental knowledge for age dependent intrinsic network structural changes as a reference for pathological aging studies where altered DMN activity needs to be reliably differentiated from that observed in healthy aging.

        Researcher(s)

        Research team(s)

        Project type(s)

        • Research Project

        In vivo interleukin-13 mRNA gene therapy for modulation of neuroinflammatory lesions : a pre-clinical proof-of-concept study in the cuprizone mouse model of multiple sclerosis. 01/01/2015 - 31/12/2017

        Abstract

        This project represents a formal research agreement between UA and on the other hand the client. UA provides the client research results mentioned in the title of the project under the conditions as stipulated in this contract.

        Researcher(s)

        Research team(s)

          Project type(s)

          • Research Project

          Image-guided decoding of mechanisms involved in healthy, accelerated and pathological aging. 01/10/2014 - 30/09/2017

          Abstract

          Aging has profound effects on many cellular processes that predispose to neurodegeneration, impairment in cognitive function, as well as changes in brain functional connectivity networks (e.g. default mode network) and synaptic alterations. However, the key mechanisms orchestrating brain aging remain largely unknown. More and more findings in rodents and humans have established that inflammatory processes in the hypothalamus can contribute to neurodegeneration upon aging via reproductive (HPG) axis. However, the exact mechanisms by which (i) Inflammatory signalling in the hypothalamus contributes to the occurrence of age-related functional connectivity and synaptic alterations, and (ii) hypothalamic HPG signalling modulates age-related neurodegeneration and cognitive changes are not well understood and need further investigation. The main goals of this project are to investigate: (i) how deregulation of the HPG axis impacts brain networks that display aging decline, (ii) how hypothalamic inflammation is steering deregulation of HPG axis in healthy aging, accelerated aging and pathological aging, and (iii) how hypothalamic inflammatory responses become activated upon healthy, accelerated and pathological aging, with specific focus on cellular, connectional architecture of functional networks. This project will contribute new information that will greatly increase our understanding about underlying mechanisms of hypothalamus-driven systematic aging of the brain.

          Researcher(s)

          Research team(s)

          Project type(s)

          • Research Project

          Unravelling the role of neuroglobin in neuroprotection and neuroplasticity. 01/10/2014 - 30/09/2016

          Abstract

          The general objective is to obtain insight in the contribution and working mechanism of two vital processes in the brain: 1) neuroprotection and 2) 17β-estradiol (E2) induced neuroplasticity. As we aim to focus on different expression patterns of Ngb both in vitro and in vivo, the first objective is to improve valid Ngb overexpressing and knock-out (KO) systems.

          Researcher(s)

          Research team(s)

            Project type(s)

            • Research Project

            Postdoctoral research dr. Prim Singh. 01/12/2013 - 30/03/2014

            Abstract

            This project represents a formal research agreement between UA and on the other hand BBSRC. UA provides BBSRC research results mentioned in the title of the project under the conditions as stipulated in this contract.

            Researcher(s)

            Research team(s)

              Project type(s)

              • Research Project

              Improving efficacy of peripheral anti-glioma vaccination by local modulation of the tumour's immunosuppressive microclimate. 01/10/2013 - 30/09/2015

              Abstract

              In this doctoral research proposal, we propose to combine both innate and adaptive immune stimulation approaches in order to mount a more potent anti-tumour response. In addition, an in-depth characterisation of central nervous system and peripheral immune changes will be performed following tumour induction and/or anti-tumour vaccination. With this research project we therefore aim to contribute to a better understanding of cellular interactions in the events of tumour growth and eradication, and to the improvement of current immunotherapeutic interventions of GBM.

              Researcher(s)

              Research team(s)

                Project type(s)

                • Research Project

                Transgenic reporter animal collection for experimental research (TRACER). 06/06/2012 - 31/12/2013

                Abstract

                TRACER: Transgenic Reporter Animal Collection for Experimental Research. Tracer will consist of a widespread variety of transgenic reporter mice, which directly fit within the research aims of two (bio)medical frontline research domains of the University of Antwerp, namely "Neurosciences" and "Imaging".

                Researcher(s)

                Research team(s)

                  Project type(s)

                  • Research Project

                  Functional identification of innate immune responses following stem cell implantation in the central nervous system of mice. 01/10/2011 - 10/04/2013

                  Abstract

                  In this PhD project we aim to completely characterize the migration behaviour, fate and physiology of different stem cell types following administration in the 'experimental autoimmune encephalomyelitis' (EAE) mouse model for Multiple Sclerosis. A combination of in vivo (BLI and MRI) and post-mortem (histology) imaging modalities will be used to reveal different cell characteristics and compare these characteristics between the different cell types included in this study. These results will enable us to effectively modify stem cell populations in order to enhance possible therapeutic effects (e.g. enhanced migration towards target sites and increased survival of grafted stem cells).

                  Researcher(s)

                  Research team(s)

                  Project type(s)

                  • Research Project

                  Neural Stem Cells: molecular and physiological control of in vivo migration and differentiation. 01/01/2011 - 31/12/2014

                  Abstract

                  This proposed multidisciplinary research consortium, consisting of 6 different laboratories from the University of Antwerp, aims to understand the cellular and/or functional interactions of NSC implants in healthy and injured neural tissue (cuprizone-mediated demyelinisation mouse model). With this research project, which focuses on the in vivo molecular and physiological control of NSC, we aim to contribute to the in vivo study and modulation of NSC migration, survival, differentiation and functional integration.

                  Researcher(s)

                  Research team(s)

                    Project type(s)

                    • Research Project

                    Immunomodulatory therapy for spinal cord injury via IL-4 en IL-13 secreting stem cells. 01/01/2011 - 31/12/2014

                    Abstract

                    The aim of this research project is to investigate the effect of IL-4 and IL-13 delivered by locally applied autologous neural stem cells (NSC) on axon regeneration after traumatic CNS injury using both in vitro as well as in vivo models.

                    Researcher(s)

                    Research team(s)

                      Project type(s)

                      • Research Project

                      Control of glioblastoma by modulation of the brain's innate immune responses. 01/01/2011 - 31/12/2014

                      Abstract

                      We first aim to identify and functionally describe immune-suppressive proteins and/or signalling molecules on glioma cells, which lead to inhibition of microglia. Then, we will develop strategies to modify microglia in order to prevent inhibition by glioma cells in vitro and in vivo. We specifically aim to genetically engineer microglia with 'short interfering RNAs' against receptors or signalling molecules involved in immune inhibition by GL261 cells in order to improve their cellular therapeutic potency.

                      Researcher(s)

                      Research team(s)

                        Project type(s)

                        • Research Project

                        Characterisation of innate immune responses in the central nervous system: modulation towards immunological acceptance of allogeneic cellular grafts. 01/01/2011 - 31/12/2014

                        Abstract

                        In this project, we aim to further elucidate the mechanisms leading to immune-mediated rejection of allografts in the CNS. For this, we will non-invasively (by in vivo bioluminescence imaging) identify the exact timing and degree of microglia immune-reactivity in relation to immune-mediated rejection of different allogeneic adult-, embryo- and placenta-derived cell populations.

                        Researcher(s)

                        Research team(s)

                          Project type(s)

                          • Research Project

                          'Molecular Imaging' meets 'Imaging Molecules' 01/07/2010 - 30/06/2014

                          Abstract

                          Magnetic Resonance imaging plays a crucial role in stem cell research in order to investigate whether administered stem cells are able to migrate to the target organ, locally survive, differentiate and contribute to regenerated tissue. However, knowledge regarding the interaction of MRI contrast agents with (sub)cellular structures is lacking. In this project, we will use advanced TEM techniques to investigate different MRI contrast agents and loading techniques for neural stem cells.

                          Researcher(s)

                          Research team(s)

                          Project type(s)

                          • Research Project

                          Neural stem cells: molecular and physiological control of in vivo differentiation, migration and immunogenicity. 01/10/2009 - 30/09/2012

                          Abstract

                          Future research will be focussed on three main topics: 1) Characterization of neural stem cells. While culture conditions have recently been established for adherently growing NSC, many questions remain regarding: (a) optimal growth conditions, (b) identity, and (c) differentiation potential in vitro and in vivo. 2) In vivo migration and survival of neural stem cells. While NSC-based therapies have demonstrated beneficial outcome in animal models of neurotrauma and/or -inflammation, currently there is no detailed knowledge regarding in vivo survival, migration and function of transplanted NSC. 3) Immunobiology of neural stem cells. While NSC-based therapies are being developed worldwide in various animal models, currently there is no knowledge regarding the in vivo interaction between the brain's innate/adaptive immune system and allogeneic NSC implants.

                          Researcher(s)

                          Research team(s)

                            Project type(s)

                            • Research Project

                            Monitoring and modulation of stem cell migration towards brain and spinal cord lesions in a murine model for experimental autoimmune encephalomyelitis. 01/10/2009 - 30/09/2011

                            Abstract

                            This is a fundamental research project financed by the Research Foundation - Flanders (FWO). The project was subsidized after selection by the FWO-expert panel.

                            Researcher(s)

                            Research team(s)

                            Project type(s)

                            • Research Project

                            Clinical and preclinical research of the effect of cellular mediators on the modulation of pathogenic responses in multiple sclerosis. 01/01/2009 - 31/12/2012

                            Abstract

                            In this project, we want to further investigate and exploit the capacity of DC and Treg to correct or modulate pathogenic responses in MS patients. Current research will provide the foundation for the eventual development of a cellular vaccine for the treatment of MS. Depending on the results of this study it can be envisaged to treat patients suffering from MS with tolerogenic DC and/or immunosuppressive Treg in order to eliminate or inactivate autoreactive T cells.

                            Researcher(s)

                            Research team(s)

                              Project type(s)

                              • Research Project

                              Characterization of immune responses against allogeneic stem cell implants in the central nervous system. 01/01/2009 - 31/12/2011

                              Abstract

                              In this project, we will investigate the role of innate (microglia) and adaptive (T-cells) immune responses leading to immunological rejection of allogeneic stem cell implants in the central nervous system of immunocompetent mice.

                              Researcher(s)

                              Research team(s)

                                Project type(s)

                                • Research Project

                                Development of combined magnetic resonance imaging and bioluminescence imaging to study stem cell migration and survival in mouse brain after neurotrauma. 01/10/2008 - 30/09/2009

                                Abstract

                                In this study, we aim to investigate whether genetic modification of exogenous stem cells with chemokine receptors enhances their migration towards lesions in brain tissue. Magnetic resonance imaging (MRI) and bioluminescence imaging (BLI) technology will be used and validated to visualize migration and survival of grafted stem cells in vivo.

                                Researcher(s)

                                Research team(s)

                                Project type(s)

                                • Research Project

                                CRYo-banking of stem cells for human therapeutic application. (CRYSTAL) 01/02/2007 - 31/01/2010

                                Abstract

                                In this EU project, partners UA (Antwerp) and KUL (Leuven) will investigate whether transient genetic modification of cord blood and bone marrow hematopoïetic stem cells with mRNAs encoding transcription factors, growth factors and receptors (all involved in self-renewal and migration of stem cells) results in improved in vivo repopulation of SCID mice. This research will lead to improved methodology for hematopoietic stem cell transplantation in adults and children.

                                Researcher(s)

                                Research team(s)

                                  Project type(s)

                                  • Research Project

                                  Development of reporter gene imaging for MRI and BLI assessment of migration and survival of transplanted mesenchymal and neural stem cells after traumatic brain injury in mice. 01/10/2006 - 30/09/2010

                                  Abstract

                                  Stem cell transplantation after neurotrauma is a promising field of research in current biomedical research. However, to date, little is known about successful migration and survival of transplanted stem cell populations on site of trauma. This project aims to develop genetically modified adult and embryonic stem cell populations which can be assessed by MRI and BLI after transplantation in traumatised mouse brain.

                                  Researcher(s)

                                  Research team(s)

                                  Project type(s)

                                  • Research Project

                                  Transplantation of embryonic stem cell-derived neural stem cells after spinal cord and traumatic brain injury. 01/10/2006 - 30/09/2009

                                  Abstract

                                  This project will investigate whether transplantation of defined embryonic stem cell-derived neural stem cells (ES-NSC), genetically modified to secrete neurotrophic factors, can support or improve recovery after TBI and SCI. An improved recovery can be due to: A) a decreased secondary neural loss due to secretion of neurotrophic factors, and/or B) an enhanced neural recovery due to functional integration of transplanted ES-NSC and/or recruited endogenous NSC.

                                  Researcher(s)

                                  Research team(s)

                                    Project type(s)

                                    • Research Project

                                    Exploring the functional and histological recovery of damaged neural tissue after traumatic spinal cord injury in rat: modulation by in vivo transplantation of in vitro transfected adult mesenchymal and embryonic stem cells. 01/10/2006 - 30/09/2007

                                    Abstract

                                    Researcher(s)

                                    Research team(s)

                                    Project type(s)

                                    • Research Project

                                    Modulation of immune responses directed against allogeneic cell transplants in mouse brain . 01/07/2006 - 31/12/2010

                                    Abstract

                                    Transplantation of embryonic stem (ES) cell-derived neural progenitor cells (ES-NPC) will become an important option for treatment of neurological trauma . However, due to technical limitations, such transplantations will most likely be using NPC derived from allogeneic ES cells. This project investigates different immunological strategies in order to overcome rejection of transplanted murine ES-NPC in brain of MHC-mismatched mice.

                                    Researcher(s)

                                    Research team(s)

                                      Project type(s)

                                      • Research Project

                                      Transplantation of adult and embryonic stem cells genetically modified with neurotrophic factors after traumatic brain injury in mice. 01/05/2005 - 31/12/2006

                                      Abstract

                                      Neurological trauma after sport, work or traffic accidents frequently lead to morbidity and mortality among adults and children. A promising therapy for neurological injury is stem cell transplantation. This project will investigate whether transplantation of adult and embryonic stem cells, genetically modified with neurotropic factors, might improve recovery after traumatic brain injury in mice.

                                      Researcher(s)

                                      Research team(s)

                                        Project type(s)

                                        • Research Project

                                        Exploring the functional and histological recovery of damaged neural tissue after traumatic spinal cord and brain injury in mice : modulation by in vivo implantation of in vitro transfected embryonic stem cells. 01/10/2004 - 30/09/2006

                                        Abstract

                                        Currently, there is no effective therapeutical approach for central nervous system injuries (both brain and spinal cord injury). Our research project will focus on 1) the tranplantation of embryonic stem cell-derived neural cell types in order to replace the damaged neural tissue, and 2) the transplantation of adult stem cells genetically modified in order to produce neurotrofic factors for regeneration of damaged neural tissue. Our project will be divided into two parts: a spinal cord injury model and a traumatic brain injury model. For the spinal cord injury model, a surgical transsection will be made on the spinal cord of rat, followed by tranplantation of both non- and gene-modified adult/embryonic stem cells at different time points in the injured spinal cord. Functional recovery will be monitored using the BBB-score for evaluation of locomotor function. For the brain injury model, induced via a weight impaction on the skull of the mouse, both non- and gene-modified adult/embryonic stem cells will be transplanted at different time points in the injured brain. Functional recovery will be monitored using standarised cognitive and motoric tests

                                        Researcher(s)

                                        Research team(s)

                                        Project type(s)

                                        • Research Project

                                        Differentiation of embryonic stem cells into neural cell types following electroporation with mRNA encoding neural key regulatory genes. 01/01/2004 - 31/12/2005

                                        Abstract

                                        Despite new surgical techniques, pharmacological treatments and functional neurological stimulation methods, brain and spinal cord injuries remain a large medical and social problem. Therefore novel research aims at the development of new therapies reaching further than current revalidation strategies. Stem cell research offers promising opportunities for the development of new methods in order to repair or replace damaged tissues or cells. Stem cells have the potential to differentiate towards a neural phenotype, creating the possibility to replace cerebral or spinal neurons, astrocytes and oligodendrocytes. The goal of this project is to investigate whether embryonic stem cells, both mouse and human, genetically loaded with mRNA encoding neural key regulatory genes can differentiate towards neural cell types in vitro and in vivo (mouse model).

                                        Researcher(s)

                                        Research team(s)

                                          Project type(s)

                                          • Research Project

                                          Use of cultured human dendritic cells loaded with defined or full spectrum tumor antigens for optimal stimulation of in vitro anti-tumor immunity. 01/10/2001 - 30/09/2003

                                          Abstract

                                          Although tumor-specific antigens have been described for a number of cancers, the immune response to these antigens is often inexistent or deficient. One possible explanation for this deficient immune response is that tumor cells themselves do not function adequately as antigen presenting cells (APC). Therefore, gene transfer in dendritic cells (DC), which are very potent antigen presenting cells of the immune system, could be a useful strategy for tumor immunotherapy. In vitro cultured human dendritic cells will be transfected with mRNA encoding tumorassociated/specific geneproducts or uploaded with the full antigenic spectrum of tumorcells (tumorextracts, cellysates or total RNA). After cocultivation with autologeous CD8+ T-ceIls we will investigate if the cultured cytotoxic T-cells show a direct speci-ficity against tumorcells. The main purpose of these experiments is the development of a DC-based in vitro immunotherapy protocol for coloncancer, cervixcancer and lymphomas.

                                          Researcher(s)

                                          Research team(s)

                                            Project type(s)

                                            • Research Project

                                            Use of cultured human dendritic cells loaded with defined or full spectrum tumor antigens for optimal stimulation of in vitro anti-tumor immunity. 01/10/1999 - 30/09/2001

                                            Abstract

                                            Although tumor-specific antigens have been described for a number of cancers, the immune response to these antigens is often inexistent or deficient. One possible explanation for this deficient immune response is that tumor cells themselves do not function adequately as antigen presenting cells (APC). Therefore, gene transfer in dendritic cells (DC), which are very potent antigen presenting cells of the immune system, could be a useful strategy for tumor immunotherapy. In vitro cultured human dendritic cells will be transfected with mRNA encoding tumorassociated/specific geneproducts or uploaded with the full antigenic spectrum of tumorcells (tumorextracts, cellysates or total RNA). After cocultivation with autologeous CD8+ T-ceIls we will investigate if the cultured cytotoxic T-cells show a direct speci-ficity against tumorcells. The main purpose of these experiments is the development of a DC-based in vitro immunotherapy protocol for coloncancer, cervixcancer and lymphomas.

                                            Researcher(s)

                                            Research team(s)

                                              Project type(s)

                                              • Research Project