Abstracts from the 12th meeting of the International Society for Neural Transplantation and Restoration

نویسندگان

  • Arturo Alvarez-Buylla
  • Luis Azmitia
  • Philipp Capetian
  • Mariana Klett
  • Máté Döbrössy
  • Guido Nikkhah
چکیده

s from the 12th meeting of the International Society for Neural Transplantation and Restoration NeuroReport 2014, 25:139–163 Note from the Publisher The following abstracts section presents abstracts from all presenters at the 12th meeting of the International Society for Neural Transplantation and Restoration. They were meant to be included in last year’s December issue of NeuroReport, but unfortunately were omitted. The Publisher and Editors apologize to readers and provide them here in the February 2014 issue. ABSTRACTS OF ORAL PRESENTATIONS Repairing neural circuits with local circuit neuronsS OF ORAL PRESENTATIONS Repairing neural circuits with local circuit neurons Arturo Alvarez-Buylla San Francisco, CA, USA Cortical GABAergic interneurons originate in the subpalial ganglionic eminences (GE). Their integration into the forebrain and cortical circuits is essential to achieve proper inhibitory-excitatory balance and appears to also be key for the induction of cortical plasticity. Importantly, these cells retain their long-range migratory potential and can integrate into neural circuits when grafted into the postnatal and adult brain. Remarkably, their numbers are not adjusted by exogenous signals, but internally in a cell or population autonomous manner. This allows for the addition by transplantation of numerous interneurons to cortex that disperse and functionally integrate. These grafted cells can reinitiate periods of cortical plasticity and can repair imbalances in animal models of epilepsy. Cortical interneuron transplantation is likely to have a wider therapeutic use as we learn more about how specific subtypes of these GABAergic cells are affected in different human diseases. I will discuss recent data suggesting the large-scale contribution of caudal and probably lateral GEs to the human cortical interneuron population. Interestingly, some of these large streams of putative migrating interneurons are retained into the postnatal human brain. These findings suggest new approaches for the repair of cortical circuits. Directly reprogrammed neural precursors from patient-specific fibroblasts Luis Azmitia, Philipp Capetian, Mariana Klett, Máté Döbrössy and Guido Nikkhah Freiburg, Lübeck and Erlangen, Germany Yamanaka et al. 2007, appeared as a mile stone in cell reprogramming exploring a source of induced pluripotent stem (iPS) cells, a valuable tool and ally for cell therapy. Nevertheless, the process continued to be complicated and time consumption, involving risks of vector integrations. We combined an insertion free approach of transfecting 3 plasmids carrying the reprogramming factors (Okita et al., 2011) into fibroblasts from Huntington’s disease (HD) patients. The cells were propagated under neuralizing conditions and neural rosettes (2 weeks after transfection) were allowed to get confluent, picked and expanded; leading to a high yield of directly reprogrammed neural cells (drNCs). drNCs were positive for Nestin, BIII Tubulin, PLZF and ZO1; and evidenced Neurotensin, FoxG1, LHX9 and Pax6. Thereafter the exposure to FGF2, FGF8, Purmorphine and CHIR99020; DLX, DSH, OTX2, GAD67 and Darpp32 were confirmed. 21 weeks after transplantation, Nestin, BIII, PSA-NCAM, GFAP, Oligodendrocyte2 and Glutamate were positive with no signs of tumor formation. This model, gave rise an early developmental phenotype of stem cells (forebrain). It evidenced malleability through addition of growth factors and small molecules: rostralization of an established commitment, including Darpp32 positive cells. It highlights a valuable tool in understanding the neurodegenerative diseases and neural development. Neurotrophins: from bench to bedside Yves-Alain Barde Cardiff, UK Neurotrophins are small dimeric proteins encoded by 4 genes in mouse and human. The lack of such genes in species used by geneticists to dissect signaling pathways has seriously complicated progress in this field. Thus, only recently was it realized that 2 of the long known neurotophin tyrosine kinase receptors, TrkA and TrkC actually kill neurons in the absence of ligand-mediated activation, thus explaining the dependency for survival of some neurons on nerve growth factor established over 50 years ago [1]. In addition, the strong structural relatedness of neurotrophins and their receptors led to unwarranted extrapolations about their mode of action. In particular, nerve growth factor in the peripheral nervous system served as the dominant conceptual framework to infer how neurotrophins would work in the brain. But recent results indicate that brain-derived neurotrophic factor (BDNF), by far the most widespread neurotrophin in the brain, is localized preand not post-synaptically in the nerve terminals of excitatory neurons [2] and that it is not a This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License, where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially. Abstracts 139 0959-4965 c 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins DOI: 10.1097/01.wnr.0000443076.61006.22 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. significant survival factor for most CNS neurons [3]. This is in line with functional results obtained by others indicating that the rodent striatum is fed forward by cortical afferents delivering BDNF, a significant body of work of high relevance to diseases such as Huntington’s. This feed forward mechanism seems to be needed by many GABA-ergic neurons to reach, possibly also to keep, their normal size even if BDNF is not needed for their survival. For example, in animals lackingMecp2, the gene associated with most cases of Rett Syndrome, the levels of BDNFare markedly reduced in some brain areas, including the cerebral cortex. In young adult animals, the size of the striatum is significantly decreased in Mecp2 mutants [4] and we recently showed that the sphingosine analogue fingolimod, a drug recently introduced as the first oral treatment of multiple sclerosis, increases BDNF levels and restores the size of the striatum to a volume close to normal [4]. It is possible then that BDNF levels may modulate the growth of some brain areas even in the adult, a notion that has hardly been explored thus far, in large part because of misleading results obtained with transgenic animals overexpressing BDNF. However, recent results from our laboratory indicate that BDNF overexpression leads to the secretion of unprocessed pro-BDNF, a protein counteracting the growth effects mediated by processed BDNF. By contrast, results obtained with another mouse mutant indicate that increased levels of physiologically processed BDNF are accompanied by a progressive, massive growth of some brain areas in the adult [2,5]. These new results suggest then that while BDNFis not a significant factor for the survival of most CNS neurons, in line with finding that its receptor TrkB does not cause neuronal death in the absence of ligand [1], it may regulate the size of various brain areas, even in the adult. References 1 Nikoletopoulou V, Lickert H, Frade JM, Rencurel C, Giallonardo P, Zhang L, et al. Neurotrophin receptors TrkA and TrkC cause neuronal death while TrkB does not. Nature 2010; 467:59–63. 2 Dieni S, Matsumoto T, Dekkers M, Rauskolb S, Ionescu MS, Deogracias R, et al. BDNF and its pro-peptide are stored in presynaptic dense core vesicles in brain neurons. J Cell Biol 2012; 196:775–788. 3 Rauskolb S, Zagrebelsky M, Dreznjak A, Deogracias R, Matsumoto T, Wiese S, et al. Global deprivation of brain-derived neurotrophic factor in the CNS reveals an area-specific requirement for dendritic growth. J Neurosci 2010; 30:1739–1749. 4 Deogracias R, Yazdani M, Dekkers MP, Guy J, Ionescu MC, Vogt KE, et al. Fingolimod, a sphingosine-1 phosphate receptor modulator, increases BDNF levels and improves symptoms of a mouse model of Rett syndrome. Proc Natl Acad Sci U S A 2012; 109:14230–14235. 5 Heyden A, Ionescu MC, Romorini S, Kracht B, Ghiglieri V, Calabresi P, et al. Hippocampal enlargement in Bassoon-mutant mice is associated with enhanced neurogenesis, reduced apoptosis, and abnormal BDNF levels. Cell Tissue Res 2011; 346:11–26. TRANSEURO: what has it achieved to date? Roger A. Barker Cambridge, UK Trials of human fetal ventral mesencephalic tissue (hfVM) transplants in patients with Parkinson’s Disease (PD) have produced mixed results with some individuals showing excellent long term outcomes and others developing sideeffects without benefit. The reasons for this may relate to patient selection, tissue preparation and support (pre and post implantation) and trial design. In 2006 a working group was set up to discuss these issues which led to the successful funding of an EU-FP7 grant looking at hfVM grafts in a new trial, called TRANSEURO. Over the last 3-4 years TRANSEURO has moved to the point of starting this new trial by addressing issues in the laboratory to do with tissue preparation at a GMP level, as well as recruiting and following up a cohort of younger early stage patients using clinical and imaging assessments. In this talk I will outline the rationale and status of this new trial. Stem cell therapy for stroke: preclinical evidence and an update on FDA-approved clinical trials Cesar V. Borlongan Tampa, Florida, USA Background and purpose: Stem cells possess excellent therapeutic applications for ischemic brain injury. In this talk, I will provide scientific evidence that intravascular and intracerebral transplantation of stem cells produce behavioral and histological benefits in experimental models of stroke. I will highlight gating preclinical evidence that form the basis for translating stem cell therapy in stroke from the laboratory to the clinic. Methods: In carefully assessing the safety and efficacy of stem cell therapy for stroke, I refer to the 3 meetings of STEPS or Stem cell Therapeutics as an Emerging Paradigm for Stroke. Here, I will discuss the lab-to-clinic experimental design that is of keen interest to the academicians, physicians, NIH program directors, US FDA regulators, and biomedical company stakeholders. Results: To date, there are 9 FDA-approved clinical trials of stem cell therapy for stroke. Safety and efficacy outcomes from these trials remain to be reported. Conclusion: Stem cell therapy for stroke remains an experimental treatment, and currently being tested in limited clinical trials. The need for transparent translational guidance on how to expedite the entry of this cell therapy to the clinic is warranted to abrogate the mortality and morbidity inherent in stroke. Disclaimer: CVB holds patents and patent applications related to stem cell therapy, and serves as consultant and editor to a number of stem cell-related biotech companies and scientific journals. Funded by NIH, Celgene, and Sanbio. Cellular autonomy and neurodegeneration Siddharthan Chandran Edinburgh, UK Neurodegenerative disorders such as Alzheimer’s disease, amyotrophic lateral sclerosis (ALS) and Parkinson’s disease are characterised by the selective loss of specific neuronal 140 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. subtypes. While the precise mechanisms underlying neuronal dysfunction and subtype specificity remain the subject of intense study, the cellular environment in which degenerating neurons reside has been shown to be an integral part of the disease. In turn, until comparatively recently glial damage and death has been the focus of other diseases such as multiple sclerosis. An emerging consensus across these diseases is that, dependent on context, glia can be injurious setting the pace of, and perhaps in some instances initiating neurodegeneration or even neuroprotective. My talk will discuss glial-neuronal interaction and the opportunities human stem cell systems offer to model aspects of cellular autonomy in the context of in the context of neurodegeneration – regeneration. AAV9-mediated EPO gene delivery in a rat model of Parkinson’s disease Wei-Ming Duan, Chun Yang, Yue-Qiang Xue, Ronald L. Klein and Li-Ru Zhao Beijing, China, Shreveport, Louisiana and Syracuse, New York, USA The aims of the work were to examine cellular and behavioral effects of intrastriatal injections of adenoassociated virus serotype 9 vectors the human EPO gene (AAV9-hEPO) into the brain of 6-OHDA-lesioned rats, and to evaluate inflammatory and immune responses against AAV9-hEPO vectors in the striatum of the rats. We observed that expression of the human EPO gene was robust and stable in the striatum and the substantia nigra. As a result, nigral dopaminergic neurons were protected against 6-OHDA-induced toxicity. Amphetamineinduced rotational asymmetry and spontaneous forelimb use asymmetry were both attenuated. Intramuscular, but not intrastriatal injections of AAV9-hEPO resulted in reduced levels of hEPO transduction and increased levels of the major histocompatibility complex class I and class II antigen expression in the striatum following AAV9-hEPO re-administration. There were infiltration of the cluster of differentiation 4 (CD4)-and CD8-lymphacytes, and accumulation of activated microglial cells and astrocytes in the virally injected striatum. In addition, the sera from the rats with intramuscular injections of AAV9-hEPO contained greater levels of antibodies against both AAV9 capsid protein and hEPO protein than the other treatment groups. Acknowledgements: This work was funded by the Chinese Ministry of Science and Technology, and Beijing Natural Science Foundation. ‘‘The Land of My Fathers’’: the pioneers of neural transplantation in whose footsteps we walk Stephen B. Dunnett Cardiff, Wales, UK The Welsh national anthem reminds us that it is good to know the roots from where we come, and inspires us to build upon the past struggles of the forefathers of our field that have allowed us to get to where we are today. I remember the early attempts at cell transplantation from Paré, Thompson and Dunn to the consistent achievements of Glees and Le Gros Clark. I consider the widespread neglect of the quite remarkable results of these pioneers by the biologists of the day in the light of an (over-rigid) interpretation of Cajal’s dictum that all regeneration in the adult mammalian central nervous system is invariably abortive. That zeitgeist was only convincingly challenged with the synchronous studies of Raisman, Altman and Olson at the turn of the 1970s using novel anatomical techniques for ultrastructural pathway tracing, thymidine birthdating, and intraocular transplantation, respectively. Using the new methods of catecholamine fluorescence to visualise specific populations of central and peripheral neurons and their connections, two schools rapidly emerged in central and southern Sweden under the leaderships and inspiration of Lars Olson at the Karolinska and Anders Björklund in Lund, respectively, that defined the conditions for cell transplantation in the adult mammalian brain. The first wave of studies using these new techniques sought to characterise the principles of cell survival, axon growth, targeting and formation of appropriate connections in development and in regeneration using primarily anatomical tools. Then at the turn of the 1980s, a second wave of activity broadened the field to investigate the extent to which transplanted cells could have a functional impact on the behaviours of the host animal, opening the way to development of reparative cell based therapies. A first set of pioneering studies demonstrated functional recovery of motor symptoms in rats with dopamine-depleting lesions following embryonic mesencephalic transplants to effect dopamine-cell replacement, and led swiftly (some might think excessively so) to the first clinical trials in patients of adrenal cell transplants. These observations stimulated a proliferation of applications in other model systems, which I illustrate by the studies of (i) John Sladek and colleagues on neuroendocrine function by hypothalamic repair in the diabetes insipidus Brattleboro rat; (ii) Albert Aguayo and the McGill team on peripheral nerve bridges to allow long distance central axon growth across the site of injury in spinal cord; and (iii) Ray Lund and team on restoration of retinal tectal reconnections. These latter studies included the first indication that rehabilitation is essential in conjunction with structural repair to maximise functional recovery, and provide the foundations for the first in man clinical trials of pluripotent and stem cell transplantation in macular degeneration, presented elsewhere in this international symposium. These foundations underpin the explosion of use of transplantation technologies over the last two decades both for the analysis of fundamental neurobiology of brain development, plasticity and reorganistaion, as well as providing the basis for the rapid growth of cell therapy as a leading strategy in contemporary regenerative medicine. Abstracts 141 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Finally, I remember the history of the INTR symposia themselves, the first of which was held in Lund in 1984 and which have continued on a regular 2-3 year cycle ever since. The INTR symposia have established and continue to foster a rare spirit of collegiality and collaboration in our field of neural transplantation research, as well as having spawned the regional societies of NECTAR and ASNTR in Europe and North America respectively. We look forward with great optimism to the 13 International Symposium to be held in Beijing in 2013. Reference 1 Dunnett SB. Neural transplantation. History of Neuroscience, Handbook of Clinical Neurology; 2010. pp. 885–912. Targeting the extracellular matrix to repair the damaged nervous system James W. Fawcett Cambridge, UK Repair of damage to the nervous system requires axon regeneration, plasticity and cell replacement. The extracellular matrix plays a central role in restricting these processes, mainly through the action of chondroitin sulphate proteoglycans (CSPGs). An important agent in removing this restriction has been the enzyme chondroitinase, which removes glycosaminoglycan (GAG) chains from CSPGs. CSPGs are up-regulated in glial scar tissue around injuries to restrict axon regeneration, which can be enhanced with chondroitinase. However the positive effect of chondroitinase on functional recovery is mostly through reactivation of plasticity. Chondroitinase treatment enhances recovery from many forms of CNS damage, including spinal cord injury, where the reactivation of plasticity enables successful rehabilitation. Chondroitinase also has profound effects on memory, prolonging object memory in normal animals and restoring memory in an Alzheimer’s model. CPSGs control plasticity mainly through their participation in perineuronal nets (PNNs), cartilage-like structures surrounding neurons, which appear as critical periods for plasticity close. PNNs contain inhibitory CSPGs, hyaluronan, link protein and tenascin-R, partly produced by the neurones themselves and partly by surrounding glial cells. All neurones with PNNs express both a hyaluronan synthase enzyme and a link protein, and these are the key components that trigger the formation of the structures. Link protein knockout animals lack normal PNNs on their dendrites, and these animals retain plasticity into adulthood, and show prolongation of memory identically to animals treated with chondroitinase. The action of the CSPGs is due to their sulphated GAGs. In the CNS these bind to and localise Semaphorin3A and OTX2 to PNNs. OTX2 is involved in the maturation of inhibitory interneurons, while Semaphorin3A is an effector of the PNNs involved in control of plasticity. Grafting of rat and porcine fetal neuronal cells into the subthalamic nucleus in experimental epilepsy M. Gernert, B. Petersen, L. Gey, A. Handreck, W. Löscher, H. Niemann and B. Backofen-Wehrhahn Hannover, Germany and Mariensee, Germany Neuronal transplantation into brain regions involved in seizure generation or propagation is a promising experimental approach to treat drug-resistant epilepsies. We recently showed that the subthalamic nucleus (STN) might be a promising target region in this respect (Bröer et al. 2012, Neurobiol Dis. 46: 362–376). We now tested two types of rat and porcine fetal stem cells (RNSCs and PNSCs) isolated from: (A) medial ganglionic eminence (MGE), and (B) mesencephalon. Grafts of both cell types cultivated as neurospheres were inserted bilaterally into the STN (80000 cells per side). Anticonvulsant efficacy was evaluated by testing seizure thresholds before and at different timepoints after grafting (from 10 days up to 3 months). RNSCs and PNSCs derived from the mesencephalon, but not from the MGE, showed clear anticonvulsant effects ten days and moderate effects 3 months after grafting. PNSCs differentiated into neurons and astrocytes and showed expression of the GABA-synthesizing enzyme glutamic acid decarboxylase. These data indicate that grafting of neuronal mesencephalic stem cells into the STN could be a promising approach to treat epilepsy. Future work will focus on improving duration of anticonvulsant efficacy after grafting into the STN. Acknowledgements: Supported by the DFG (Ge1103/7-2). GDNF – Back in the clinic Steven S. Gill, Neil Barque, Alison Bienemann, Max Woolly, Nik Patel, Matthias Luz, Lyn Barclay, Eric Mohr, Lucy Mooney, Elizabeth Coulthard, Andrew Lawrence, Chis Marshall and Alan L. Whone Bristol, UK, Victoria, BC, Canada and Cardiff, UK Objective: To evaluate in PD the efficacy and safety of intermittent GDNF intra-putaminal infusions administered by Convection Enhanced Delivery. Background: In animal models of PD and in open label studies in PD patients, continuous intra-putaminal GDNF infusions have been shown to improve motor symptoms and, as assessed by 18F-dopa PET and post-mortem assessments, restore dopamine terminals. However, in 2003, a placebo-controlled multi-centre trial reported failure to demonstrate clinical benefit, despite improvements in PET end-points. In addition, a concurrent study in monkeys raised questions over safety. Since that point no further human investigations have been performed. In significant part, we believe, the above issues were due to a failure in the way GDNF was surgically delivered. We have now developed an in-house device which animal model studies suggest will allow GDNF to be given much more 142 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. reliably to the putamen. We feel this allows for definitive testing of GDNFs effects in humans. Methods: A phase II, single centre, randomized, double blind, placebo-controlled trial, in idiopathic PD (n=42), of intermittent bilateral posterior putamen GDNF infusions administered via Convection Enhanced Delivery (CED) was commenced in 2012. The drug delivery system comprises 4 micro-catheters, with in-line filters, and a skull mounted transcutaneous drug delivery port with 4 independent channels. 4 programmable pumps are connected to the port via an administration set on a 2 or 4 weekly basis for intermittent infusions of GDNF or placebo (aCSF) through the course of this trial. Results: The design of this 3 phase trial: Pilot Stage; Primary Study stage; and Roll-Over Stage will be discussed as well as describing the technological challenges of achieving reliable intermittent CED of GDNFover the long term and how we are attempting to address these. Conclusion: This will be the first clinical study of intermittent GDNF infusions delivered by CED in man. Generation of human neural progenitor cells from induced pluripotent stem cells that survive, migrate and integrate into the rodent spinal cord Geneviève Gowing, Dhruv Sareen, Anais Sahabian, Kevin Staggenborg, Renée Paradis, Pablo Avalos, Jessica Latter, Loren Ornelas, Leslie Garcia and Clive N. Svendsen Los Angeles, CA, US Transplantation of human neural progenitors (hNPC) derived from pluripotent stem cells (PSC’s) is a promising therapeutic strategy that has the potential to replace lost cells, modulate the injury environment and create a permissive milieu for the protection or regeneration of host neurons in disease., Here we use a novel chopping technique to isolate and expand EZ spheres from PSC’s which were then driven to an expandable iNPC spinal cord phenotype using a combination of retinoic acid followed by the mitogens EGF and FGF-2. iNPCs grown in suspension showed similar characteristics to hNPCs derived from human fetal tissues, although iNPCs grown as an adherent culture did not. Suspension iNPCs were easy to maintain using the chopping method of expansion and survived grafting into the spinal cord of athymic nude rats with no signs of overgrowth, again with very similar profiles to hNPCs derived from fetal tissues. These results suggest that iPSC-derived NPC’s could be a favorable alternative to fetal hNPCs for cellular regenerative therapies of CNS diseases. Modulating endogenous stem cells to restore learning and memory in Temporal Lobe Epilepsy William P. Gray Cardiff, Wales, UK Neurogenesis, the production of new neurons, is a restricted event in the adult human brain, largely confined to the dentate gyrus where it is important for hippocampal learning and memory. While early studies showed conflicting roles for neurogenesis in spatial learning & memory, more recent work using paradigms that involve a higher cognitive demand, have demonstrated roles for neurogenesis in both the acquisition and retrieval of spatial relational memory. Altered neurogenesis also appears to play a role in anxiety and depression. Interestingly antidepressants increase hippocampal neurogenesis, an effect that appears necessary for their behavioural effects on mood and anxiety. Hippocampal neurogenesis is affected by a myriad of factors from neural activity (learning), exercise, drugs, metabolism, the immune system as well as brain injury and disease. Altered neurogenesis may thus be an important mechanism, underlying cognitive dysfunction and altered mood across a wide variety of neurological diseases, and as such presents an attractive target for medical and surgical interventions. I will present on overview of hippocampal neurogenesis, followed by an analysis of how it is affected by temporal lobe epilepsy, and emerging strategies from our lab and others, on how it may be restored. The origins and future of pluripotency and cellular reprogramming John B. Gurdon Cambridge, UK The different cell types that compose our bodies are remarkably stable. Hardly ever do we find skin cells in the brain or liver cells in the heart. In those very special cases where some regeneration can take place in vertebrates, there is little if any evidence for a switch in cell-type. Nevertheless, nuclear transfer, cell fusion, and induced pluripotency can result in pluripotent embryo cells being derived from specialized adult cells. The mechanisms by which nuclear reprogramming can occur in these cases is beginning to be understood. It may become possible for new, regenerated cell-types to be derived from adult cells and given back to a patient so that they receive new cells of their own genetic constitution, thereby avoiding the need for immunosuppression. The history of work in this area, and the prospects for cell replacement in the future will be discussed. Imaging stem cell fate following intracerebral grafting Philippe Hantraye Fontenay-Aux-Roses, France When paving the way towards clinical applications of stem cell grafting, one crucial step is the ability to ascertain whether once implanted in a particular individual the pre-differentiated stem cells are actually evolving normally. In fact, a longitudinal, non-invasive follow-up should help us predict whether cells giving rise Abstracts 143 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. to the expected neuronal cell type or, on the contrary, turning into an unnecessary mixture of neurons and/or glial cells or even leading to an uncontrolled growth of cells that would potentially be harmful to the patient. In vivo imaging may be helpful in solving these questions provided that each of the available modalities has been carefully validated and tested for its capacity to differentiate the normal and/or the pathological fate of the cells following grafting in a living being. In addition, as these techniques have to be applied to a clinical setting, their use should meet strict regulatory and safety criteria. Of all available techniques, magnetic resonance imaging (MRI) has been long used to obtain information about graft placement, the in vivo monitoring of graft growth as well as occurrence of abnormal phenomena either associated with the grafting procedure itself (mechanical stress, hemorrhages) or the potential death of the grafted cells occurring shortly following their implantation (oedema). Besides MRI, alternative MR techniques have also been developed and studied for their potential use in stem cell research to precise the fate of these cells in the living being. In this context, H-MR spectroscopy has proven capable of providing a biochemical signature of the graft composition in vivo, which helps differentiate a graft presenting with the expected proportion of neurons and another, undoubtedly enriched in astrocytes or other glial cell types. In addition, diffusion-weighted MR imaging may also yield invaluable information on how the grafted cells extend significant processes out of their implantation sites and efficiently reconnect remote regions of the host’s brain in a normal or unexpected way. Most certainly, one of the imaging techniques that holds most promise in medical applications of stem cell research is positron emission tomography. This radioisotopic technique enables one to map and quantify the concentrations of any molecule previously labeled with a positron-emitting isotope. Depending on the radiotracer being selected for the study, PET can either serve as a means to detect adverse effects like neuroinflammation and cell rejection or to study the phenotypic differentiation of the graft using markers that are specific of the expected neuronal cell population. Metabolic markers like F-fluorodeoxyglucose can also help differentiate between surviving neurons and dying cells. Reuptake blockers like the cocaine analog F-LBT999 can also evidence the regrowth of presynaptic terminals, innervating de novo the grafted stem cells neo-differentiated into striatal GABAergic neurons. Very similarly, specific radiomarkers of the preand post-synaptic elements of the dopaminergic synapse can also be used to establish the functional status and the degree of integration of stem cells programmed to differentiate into a dopaminergic phenotype. When preparing a clinical trial, it is certain that a combination rather than the sole use of one of these imaging approaches has to be envisioned. Preclinical trials have also shown that the best combination of imaging techniques/markers may vary greatly in function of stem cell type to be used. Examples of the use of these various imaging techniques will be provided and their advantages and disadvantages will be discussed in the context of the preclinical application of stem cells for Huntington’s and Parkinson’s diseases. Global clinical neurorestoration in complete chronic spinal cord injury Hongyun Huang, Tiansheng Sun, Lin Chen, Milan Dimitrijevic, Gustavo Moviglia, Elena Chernykh, Klaus von Wild, Haluk Deda, Kyung-Sun Kang, Anand Kumar, Sang Ryong Jeon, Shaocheng Zhang, Giorgio Brunelli, Albert Bohbot, Maria Dolors, Jianjun Li, Alexandre Fogaça Cristante, Haitao Xi, Gelu Onose, Helmut Kern, Ugo Carraro, Hooshang Saberi, Hari Shanker Sharma, Alok Sharma, Xijing He, Dafin Muresanu, Shiqing Feng, Ali Otom, Dajue Wang, Koichi Iwatsu and (all authors from 19 countries) Beijing, China It is still popular in medical community that there are no any effective therapeutic methods to restore neurological functions for lesion. Though the treatment to recover function in people suffering complete spinal cord injury (SCI) remains a big challenge for clinical physicians, accumulating data have shown that partial clinical neurorestoration is possible by using cell therapy, neurostimulation or neuromodulation, neuroprosthesis or related advanced assistive devices, neurotization or nerve bridging, neurorehabilitation. Here we summarize the literatures that demonstrate patients with chronic complete SCI have been able to obtain some clinical neurorestoration, which is based on the scientific information available till April of 2013, and we discuss several important issues about these evidences. The goal of this review is to show the objective ‘‘yes or no’’ evidences of clinical neurorestoration for chronic complete SCI, which will make more people know real progresses in this field. And we hope the medical community is able to highlight the value of clinical neurorestorative treatments for CNS incurable diseases. Very long-term clinical outcome of fetal cell transplantation for Parkinson’s disease Zinovia Kefalopoulou, Marios Politis, Paola Piccini, Niccolo Mencacci, Kailash Bhatia, Marjan Jahanshahi, Håkan Widner, Stig Rehncrona, Patrik Brundin, Anders Björklund, Olle Lindvall, Patricia Limousin, Niall Quinn and Thomas Foltynie London, UK, Lund, Sweden and Grand Rapids, Michigan, USA Recent advances in stem cell technologies have renewed an interest in the use of cell replacement strategies for patients with Parkinson’s disease (PD). This study reports the very long-term clinical outcomes of fetal cell transplantation in two patients with PD. Such long-term follow-up data can 144 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. usefully inform on the potential efficacy of this approach, as well as the design of trials for its further evaluation. Two patients received intrastriatal grafts of human fetal ventral mesencephalic tissue, rich in dopaminergic neuroblasts, as restorative treatment for their PD. To evaluate the long-term efficacy of the grafts, clinical assessments were performed 18 and 15 years post-transplantation. Motor improvements developed and increased gradually over the first post-operative years, and are sustained up to 18 years post-transplantation. Despite each patient having nearly 30-years of PD with troublesome fluctuations and dyskinesias prior to transplantation, both patients now present with mild symptoms, and are independent of any pharmacological dopaminergic treatment for more than 10 years. Both patients have developed mild/moderate graftinduced dyskinesias. The results from these two cases indicate that dopaminergic cell transplantation can offer very long-term symptomatic relief in carefully selected PD patients, and provide proof-of-concept support for future clinical trials using fetal or stem cell therapies. Acknowledgements: This work was supported in part by the European Commission under the 7th Framework Programme – HEALTH – Collaborative Project Transeuro (Contract n 242003). Anti-amyloid precursor protein antibodies as a potential therapy for Alzheimer’s disease Emma J. Kidd, Martha Hvoslef-Eide and Rhian S. Thomas Cardiff, UK Cleavage of amyloid precursor protein (APP) by band g-secretases results in amyloid-b (Ab) production in Alzheimer’s disease. We raised two antibodies to the b-secretase cleavage site in APP. Our hypothesis was that these antibodies would reduce Ab levels via steric hindrance of b-secretase. Cells were incubated with various antibodies and APP, its cleavage fragments and the affinities of the antibodies for binding to the cleavage site and likely epitopes were measured by ELISAs. The ability of antibodies to enter cells was examined by immunocytochemistry. Block of b–secretase by steric hindrance was investigated. Both antibodies significantly reduced Ab levels with 2B3 being more effective than 2B12. 2B3 had higher affinity for the b–secretase cleavage site than 2B12 and bound across the cleavage site while 2B12 bound upstream. Both antibodies entered living cells and inhibited the action of b–secretase. 2B3 is a more potent antibody than 2B12 to reduce Ab levels in agreement with its higher affinity for the cleavage site and epitope location. We have proved our hypothesis that these antibodies block the b-secretase cleavage site. 2B3 is being considered as a therapeutic antibody for Alzheimer’s disease. Acknowledgements:This work was funded by the Alzheimer’s Society. Human ventral mesencephalic grafts alleviate both motor and non-motor dysfunctions in a rodent model of PD Mariah J. Lelos, Claire M. Kelly, Eduardo M. Torres, Anne E. Rosser and Stephen B. Dunnett Cardiff University, UK Although it remains the current ‘gold standard’ cell replacement therapy, characterisation of the functional efficacy of human primary tissue is currently limited to the alleviation of simple motor deficits. Given that Parkinson’s disease patients also develop impairments in a range of non-motor domains, including neuropsychiatric and cognitive deficits, it is critical to evaluate the cell therapies in terms of their impact upon these non-motor dysfunctions. To address this, we pre-trained rats on a lateralised reaction-time task, before they received a unilateral 6OHDA MFB lesion. A subset of lesion rats were then grafted with suspensions of human ventral mesencephalic tissue (hVM), of B9 weeks gestation. On both amphetamineand apomorphine-induced rotation tests, rats grafted with hVM displayed a significant reduction in rotational bias. Twelve weeks post-graft, rats were tested on two distinct versions of the operant task. Results from grafted rats revealed enhanced motor function (movement time, reaction time) as well as significant improvement of nonmotor impairments, including motivational and spatial processing (total trials completed, accuracy). These data are the first to demonstrate that VM tissue from the human fetus is capable of alleviating the non-motor dysfunctions induced by loss of striatal dopamine in Parkinson’s disease. Acknowledgements: Funded by FP7 NeuroStemCell. Directing striatal GABAergic fate specification of human pluripotent stem cells Charles Arber, Sophie V. Precious, Serafı́ Cambray, Jessica R. Risner-Janiczek, Claire Kelly, Andreas Heuer, Tristan A. Rodrı́guez, Anne E. Rosser, Stephen B. Dunnett and Meng Li Cardiff, UK The g-amino butyric acid (GABA) medium-sized spiny neurons (MSNs) are the principal projection neurons of the striatum, which specifically degenerate in the early phase of Huntington’s disease. Unlike some other neurodegenerative diseases, such as Parkinson’s, which can be effectively managed for years by dopamine-based pharmacotherapy, no effective treatment is currently available for Huntington’s. MSNs derived from human pluripotent stem cells (hPSCs) promise hope for developing transplantation-based cell therapy, studying disease aetiology and drug screening for HD. We discovered that the TGFb molecule activin can induce lateral GE (LGE)/striatal characteristics in forebrain Abstracts 145 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. neural progenitors derived from hESCs and hiPSCs. These progenitors readily differentiate into post-mitotic neurons expressing the signature marker of MSNs, dopamineand cAMP-regulated neuronal phosphoprotein (DARPP32) in culture, as well as following transplantation into the striatum of a rat model of Huntington’s disease. Significantly, grafts derived from activin A-induced striatal progenitors showed no cellular expansion and were tumour-free 16 weeks post-transplantation. Together, our findings demonstrate a novel route for directed differentiation of transplantable MSNs from hPSCs. Gene therapies for hereditary Parkinson’s disease; a strategic transition from in vivo to ex vivo Hideki Mochizuki Osaka, Japan In mouse and monkey models of Parkinson’s disease (PD), we have previously shown that intracerebral injection of adeno-associated viral vector encoding parkin, which is the causative gene of autosomal-recessive juvenile-onset PD PARK2, resulted in rescue of neuronal and behavioral phenotypes [1,2]. Delivery of parkin counteracted the attenuation of PKB/Akt pathway and protected dopaminergic neurons from apoptotic death in PD mice [2]. However, the proportion of neuronal survival was limited to a small extent and mitochondrial abnormality was not corrected in the in vivo gene therapy. Meanwhile, a recent advances on iPS cell technology led us to establishment of a novel ex vivo gene therapy for hereditary PD. We have generated dopaminergic neurons in vitro from iPS cells, derived from PARK2 patients, and found mitochondrial dysfunction and accumulation of alpha-synuclein, which resembled the histopathology in a donor patient [3]. We are studying the effects of transduction of wild-type parkin to the parkin-deficit iPS cell-derived dopaminergic neurons and transplantation of these cells in mouse and monkey models of PD to evaluate the novel ex vivo gene therapy. References 1 Yamada M, Mizuno Y, Mochizuki H. Parkin gene therapy for alphasynucleinopathy: a rat model of Parkinson’s disease. Hum Gene Ther 2005; 16:262–270. 2 Yasuda T, Hayakawa H, Nihira T, Ren YR, Nakata Y, Nagai M, et al. Parkin-mediated protection of dopaminergic neurons in a chronic MPTP-minipump mouse model of Parkinson disease. J Neuropathol Exp Neurol 2011; 70:686–697. 3 Imaizumy Y, Okada Y, Akamatsu W, Koike M, Kuzumaki N, Hayakawa H, et al. Mitochondrial dysfunction associated with increased oxidative stress and a-synuclein accumulation in PARK2 iPSC-derived neurons and postmortem brain tissue. Mol Brain 2012; 5:35. Clinical application of retinal pigment epithelium (RPE) cells derived from human embryonic stem cells Karim Ben M’Barek, Walter Habeler, Alexandra Plancheron, Ying Yang, Mohamed Jarraya, José-Alain Sahel, Marc Peschanski, Olivier Goureau and Christelle Monville Évry and Paris, France The retina is part of the central nervous system and contains specialized neurons, photoreceptors, that convert light signals into electric signals, further transmitted to the brain by different neurons. In vivo, the retinal pigmented epithelium (RPE) constitutes a distinct monolayer of pigmented cells lying between the neural retina and Bruch’s membrane, which provides essential support for the long-term preservation of retinal integrity and visual function. Given the intimate anatomical and functional relationship of RPE cells and photoreceptors, it is not surprising that the first manifestations of primary RPE disorders are problems with visions. Such is the case with age-related macular degeneration (AMD) and some forms of retinitis pigmentosa (RP) caused by mutations in genes involved in central RPE functions. RPE replacement therapies using RPE cells generated from human embryonic stem cells (hESC) provide a novel approach to a rational treatment of such forms of blindness. The aims of our study is to (i) generate a clinically-compatible polarized monolayer of RPE cells derived from human embryonic stem cells (hESC) disposed on a biocompatible membrane and (ii) test their functional properties in RP animal models in order to develop a clinical trial for patients suffering from genetic RPE disorders. The clinical grade hESC line, RC09 (Roslin Cells, Edinburgh, Scotland) is used for RPE differentiation. Our laboratory already set up and validated a reproducible differentiation protocol to generate pure RPE cells from several human pluripotent stem cells including RC-09. RPE cells obtained are polarized and functional. Our method is robust and productive: a single six-well plate of hESCs can generate 8 10 RPE cells. This protocol should now be adapted for clinically compatible conditions. Encapsulation of stem cells in an in situ gelling collagen hydrogel improves graft volume and striatal retention after intra-cerebral transplantation in the rat Ben Newland, Deirdre Hoban, Abhay Pandit and Eilı́s Dowd Galway, Ireland. Delivery of neurotrophic factors to the brain via genetically modified mesenchymal stem cells (MSCs) offers a promising neuroprotective strategy for neurodegenerative diseases. However, MSCs delivered to the CNS typically show poor survival post transplantation which limits their therapeutic efficacy. Recent studies have revealed the potential of biomaterials as supportive matrices for transplanted cells which may assist in the grafting process. In this study, an in situ gelling type I collagen hydrogel was evaluated as an intracerebral transplantation matrix for delivery of MSCs to the rat brain. We specifically investigated the effect of the hydrogel on striatal graft volume and retention. 146 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Preliminary in vitro studies confirmed that the hydrogel was neither non-toxic to MSCs seeded within it nor to neural cells seeded in juxtaposition to it. We then demonstrated that the collagen hydrogel itself was well tolerated in the rat brain, and was capable of improving the volume of the MSC graft after transplantation. Moreover, the hydrogel completely prevented ectopic deposition of MSCs along the needle track thereby increasing striatal retention of the graft. These findings indicate the potential of supportive hydrogels for intra-cerebral cell transplantation studies. Acknowledgement: This work was funded by Science Foundation Ireland. Prosavin, a dopamine gene therapy for advanced PD: phase I clinical update S. Palfi, J-M. Gurruchaga, G.S. Ralph, C. Watts, P. Buttery, S. Scorer, H. Lepetit, J. Miskin, H. Iwamuro, S. Lavisse, A. Kas, A.L. Ramelli, N. Tani, P. Dolphin, G. Fenelon, P. Brugiere, S. Kingsman, S. Naylor, R. Barker, P. Hantraye, P. Remy, P. Césaro and K. Mitrophanous Fontenay aux Roses, France, Oxford, UK, Paris, France and Cambridge, UK Parkinson’s disease (PD) is a neurodegenerative disease that results in a progressive degeneration of dopaminergic neurons. The dopamine precursor L-Dopa and dopamine agonists provide the primary standard of care and demonstrate good therapeutic benefit in the early stages of disease. However, their long term use is associated with severe motor side effects that are at least partially caused by the fluctuating nature of dopaminergic stimulation that arises from oral drug administration. As such, a therapy that provides a more continuous and local supply of dopamine to the site of pathology provides a potential approach for the development of new therapeutic strategies. ProSavin is a gene therapy product that utliises a lentiviral vector to transfer three genes that are critical for dopamine biosynthesis to the the striatum, that is depleted of dopamine in PD. Clinical evaluation of the safety and efficacy of ProSavin in mid to late stage PD patients is currently ongoing. In the study fifteen patients have received ProSavin in three dose cohorts. ProSavin has been demonstrated to be safe and well tolerated at all doses evaluated to date. There have been no serious adverse events related to Prosavin or the administration procedure and no inflammatory responses. In terms of efficacy an improvement in the primary endpoint, UPDRS Part III, has been observed in all cohorts relative to their baseline scores. Furthermore, an improvement has been maintained out to the latest timepoints evaluated to date (up to 4 years for the earliest cohort). Secondary endpoints including the patients concommitant dopaminergic medications has also demonstrated improvements. Generating neurons for use in cell therapy; challenges and possibilities Agnete Kirkeby, Ulrich Pfisterer, Jenny Nelander, Shane Grealish and Malin Parmar Lund, Sweden Parkinson’s Disease (PD) is a particularly interesting target for stem cell based therapy. The central pathology is confined to a small group of neurons in the midbrain, the nigral dopamine (DA) neurons and their projection to the striatum. Transplants of DA neurons could be used to restore DA neurotransmission in the striatum, substitute for the lost neurons, and bring back normal motor behavior. Proof-of-principle that this can work has been obtained in trials where fetal DA neuroblasts, have been transplanted to the putamen in patients with advanced PD. Despite these encouraging results, work with human fetal tissue presents a number of ethical and logistical problems and therefore does not represent a realistic therapeutic option in the future. Further progress in this field is critically dependent on the development of a bankable and renewable source of transplantable DA neurons. We have developed a method to generate human neural progenitors and neurons from human embryonic stem cells (hESCs), which recapitulates human fetal brain development. By addition of a small molecule to activate canonical WNTsignalling, we induced rapid and efficient dose-dependent specification of regionally defined neural progenitors ranging from telencephalic forebrain to posterior hindbrain fates. The DA neurons obtained via our protocol closely resembled their fetal counterparts, making them useful as a model system for studies of human fetal brain development and also for developing transplantable therapeutic cells. In parallel, we also develop cell reprogramming as an alternative source of neurons. We have found that the neural conversion genes (Mash1, Brn2a, Myt1l) can convert human fibroblasts into induced neurons (iNs). When combined with DA fate determinants, functional DA neurons can be obtained with this technique. Experimental and clinical transplantation of pluripotent stem cells for cell therapy in retinal disease Conor M. Ramsden, Michael B. Powner, Amanda-Jayne F. Carr, Matthew J.K. Smart, Lyndon da Cruz and Peter J. Coffey London, UK and Santa Barbara, CA, USA Several clinical trials of cell based therapy for retinal degeneration are currently recruiting worldwide. This clinical translation is based on over a decade of research into retinal development and physiology. One approach is to generate an appropriate replacement from stem cells, another is to use their paracrine restorative qualities. In tandem, surgical Abstracts 147 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. research into autologous retinal transplantation has evolved techniques to manipulate grafts into the subretinal space. The eye is an ideal organ for an experimental cell based therapy due to its transparency and the immune privilege conferred by the blood retina barrier. The first trials have focussed on the replacement of the retinal pigment epithelium but ultimately elements of the neural retina must be transplanted to address diseases such as advanced age related macular degeneration and retinitis pigmentosa. Medium and long term follow up of these trials is eagerly awaited. Differentiation of human pluripotent stem cells into brain-regionalized astrocytes to investigate pathogenesis of Parkinson’s and Alzheimer’s diseases Federica Rinaldi, Jennifer Badger, Lucy Crompton and Maeve Caldwell Bristol, UK Progressive neuronal degeneration and loss are hallmarks of several neurodegenerative disorders including Parkinson’s (PD) and Alzheimer disease (AD); with aetiology and pathophysiology of disease progression not yet fully elucidated. In this study we describe the generation of multipotent neural progenitor (NPs) cells from human stem cells including embryonic (hES) and induced pluripotent (iPS) cells. Our protocol faithfully recapitulates in vivo neurogenesis where neurons are generated first followed by astrocytes. We have employed RNA and protein analysis to characterize the transition from pluripotent to NP cells, and to more restricted astroglial progenitors. Ultimately, mimicking morphogen gradients occurring in vivo during neural development we have patterned NP cells to acquire basal forebrain and ventral midbrain identity thus successfully deriving brain-regionalized astrocytes. Our results reflect novel data, which have indicated the presence of heterogenic astrocytes distinguished according to their morphology, function, electrical proprieties and transcriptome. Furthermore our protocol will be of particular interest in the elucidation of the participation of regional astrocytes in neurodegenerative disorders that affect a specific brain region such as PD and AD, previously understood as exclusively neuronal disorders. Acknowledgements: This work was funded by Parkinson’s UK. Region-specific restoration of striatal synaptic plasticity by dopamine grafts in experimental parkinsonism Daniella Rylander, Vincenza Bagetta, Valentina Pendolino, Elisa Zianni, Fabrizio Gardoni, Monica Di Luca, Paolo Calabresi, Maria Angela Cenci and Barbara Picconi Lund, Sweden, Rome, Milan and Perugia, Italy Intrastriatal transplantation of dopaminergic neurons can restore dopamine levels and improve parkinsonian deficits. However, underlying mechanisms are poorly understood. Here we evaluated the synaptic plasticity in the host striatum after neural transplantation using a rat model of Parkinson’s disease. Naı̈ve rats showed distinct synaptic plasticity patterns in striatum. Ventrolateral striatum showed long-term potentiation (LTP) in approximately 63% of the neurons using the same protocol that elicited long-term depression (LTD) in the dorsolateral striatum. The LTP was linked to higher expression of post-synaptic AMPAand GluN2B NMDA subunits and was shown not to be pathway dependent. In both regions, the synaptic plasticity was abolished after dopamine-depletion and could not be restored by grafted serotonergic neurons. Solely, dopamine grafts restored the LTP and partially restored motor deficits. The restoration was only present in ventrolateral striatum where grafted reinnervation was denser compared to the dorsolateral region. These data provide a first proof of concept that dopamine transplants are able to functionally integrate into the host brain and restore neuronal deficits in striatal synaptic plasticity after experimental PD. This might have implications for the limitation in symptomatic improvement following transplantation. This work was funded by Swedish Research Council (BAGADILICO), European Community (222918, 7 framework, REPLACES), Tegger Foundation (Sweden). Clinical-grade neural progenitor cells secreting GDNF for the treatment of ALS Brandon Shelley, Genevieve Gowing, Jessica Latter, Kevin Staggenborg, Pablo Avalos, Leslie Garcia, Renee Paradis, Maximus Chen, Amanda Hurley, Jessica Zelaya and Clive Svendsen Los Angeles, USA Amyotrophic Lateral Sclerosis (ALS) is a disease in which motor neurons die due to an unknown mechanism. Human, fetal cortex-derived neural progenitor cells (hNPCs) can be expanded in vitro and survive and integrate into large and small animals following transplantation, such as rat models of ALS. Furthermore, hNPCs can be genetically modified to secrete glial cell line-derived neurotrophic factor (GDNF), a powerful growth factor that has been shown to have a neurotrophic effect in animal models. We have demonstrated that transplanting hNPCs engineered to secrete GDNF can rescue the microenvironment and preserve dying motor neurons in animal models of ALS. Moving towards the clinic, we have generated a GMPgrade master cell bank of hNPCs and sourced it to generate research and clinical-grade cell lots transduced to secrete GDNF with GMP-grade lentivirus. We have completed the dose ranging aim of the project using an ALS rat model and are moving forward with the safety/ 148 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. toxicity aim required by the FDA for approval of a phase 1/2a drug safety trial. Here we show the progress made over the first six months of the project and describe future plans, with the eventual goal of transplanting 18 patients. Acknowledgements: CIRM DR2A-05320. From Thompson to Hopkins-Dunn to Ranson; if the Pioneers could see us now! John R. Sladek Jr Aurora, CO, USA At the first of the series of twelve international meetings on neural transplantation (and now repair) near Lund in 1984, Professors Björklund and Stenevi presented a thoughtful review of the history of our field and drew attention to the findings of early investigators such as Thompson (1890), Faldino (1924), Flerko (1957) and others. At our second meeting (Rochester, 1987) we explored the great progress achieved by our modern pioneers including ongoing clinical experiments and new models of neurological diseases amenable to repair. Clearly W.G. Thompson (NYU) should be credited with the first attempt to graft brain tissue and remarkably in 1909, Walter Ranson (Northwestern Univ.) succeeded in grafting spinal ganglia and observed signs of neuronal plasticity. He described ‘‘factors’’ that must be controlling this cellular transformation, but lamented that there were no known means by which to identify them. Perhaps I’m biased, but Elizabeth Hopkins Dunn may have made the most important of these early discoveries by grafting comparatively young tissue to the adult brain and at a time when women scientists and physicians were rare and subjected to formidable obstacles to success. After graduation from the Iowa (now Grinnell) College in 1892, Dr. Dunn received her medical degree from the Northwestern University Women’s Medical School in 1894 and joined the faculty of the University of Chicago until 1906 when she moved to Woods Hole Marine Biological Laboratories, where her work extended to many other fields including publishing and cartography. On 2 January, 1914 at the 29th annual meeting of the American Association of Anatomists in Cleveland she was the only woman among 13 other scientists in her morning session and presented her findings on ‘‘Transplantation of the cerebral cortex of the albina rat’’. Her research revealed that the age of the donor tissue was critical and that neonatal tissue could survive in an adult brain. Her hand drawings of the host rat brain showing the presence of new neurons and neurites were both artistic and meaningful, and undoubtedly led to thousands of subsequent publications that utilized developing neural tissue to explore the potential for repair of neurological diseases and disorders. While so many others have made profound discoveries well beyond the scope of these early works, that Dr. Hopkins-Dunn was able to move our field forward amidst professional and likely personal obstacles suggests a brilliant mind and a persistent personality of this ‘‘First Lady of Neural Transplantation’’. Acknowledgements: Special appreciation to NIH since 1974, ASNTR since 1994 and Grinnell College, Northwestern University, New York University, and the University of Chicago for their historical insights on our early pioneers. Rab1A and Rab3B gene therapy in the Q175 knock in model of Huntington’s disease Gaynor A. Smith, Emily N. Mangano, Melissa Hayes, Jesse R. McLean, Jonathan Beagan, Sara C. Izen, Ole Isacson and Penelope J. Hallett Belmont, MA, USA Regulation of synaptic function, vesicular transport and organelle dynamics is fundamental to the normal function of the neuron. These processes are dysregulated in many neurodegenerative diseases prior to cell death, therefore providing a target for therapeutic intervention at early stages. Synaptic dysfunction and disturbances in protein transport and energy homeostasis have been reported in Huntington’s disease (HD) patients, and in experimental models with a CAG expansion in the huntingtin gene. These changes are modulated by specific direct and indirect protein interactions with huntingtin and the cytoskeleton, synapses and organelles, leading to both gain and loss of function. We have previously shown that delivery of Rab3b by gene therapy can improve neurotransmitter handling and storage capacity at presynaptic terminals, and prevent synaptic neurodegenerative changes in the dopaminergic system. We hypothesize that modulation of early pathophysiological changes by Rab proteins can enhance neuronal function in experimental models of HD. We first examined motor and cognitive functional deficits, biochemical changes, inflammation, inclusion formation, and the dysregulation of multiple proteins involved in synaptic and axonal transport, in a novel Q175 knock in (Q175 KI) mouse model of HD at 1, 6, 12 and 16 months of age. Q175 KI mice exhibited motor and cognitive deficits at 12 months, which were paralleled to striatal atrophy and microglial activation. Intracellular inclusions of mutant huntingtin were present from 6 months of age. Striatal GABA levels were increased at 1 and 6 months and a reduction in striatal dopamine observed from 6-16 months. Levels of proteins involved in synaptic and cellular transport functions were differentially altered in the striatum, motor cortex, prefrontal cortex and hippocampus at 12 months, including a reduction of the synaptic vesicle protein Rab3B, and of the ER to golgi transport protein Rab1A, in cortical regions. We are currently testing whether intra-cortical and intra-striatal gene therapy of AAV2/5 Rab1A and Rab3B in Q175 KI mice will improve secretary transport and synaptic functions of the corticostriatal pathway, thereby preventing key predegenerative changes associated with HD. Funding: CHDI Foundation. Abstracts 149 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Midkine (MDK): a newly-recognized endogenouslyproduced cytokine that, through an autocrine mechanism, is pivotal for neural induction I. Singec, A.M. Crain, B.T.D. Tobe, J. Hou, M. Talantova, K.S. Doctor, J. Choi, X. Huang, G.J. Gutierrez, D.A. Wolf, S.A. Lipton, L.M. Brill and E.Y. Snyder La Jolla, CA, USA Having devised a novel strategy for deriving pure neuroectoderm from human embryonic stem cells (hESCs), we performed the largest differential (phospho)proteomic analysis on any biological system to date, comparing hESCs with their neural stem cell (hNSC) derivatives to determine the key molecules responsible for neuralization & the transition from pluripotency to multipotency. Unexpectedly, MDK was exceptionally abundant where pluripotency marker OCT4 was receding, its expression highest in hNSCs. Though exogenous MDK promoted ‘‘neural tube’’ formation, endogenous MDK constitutively produced by the colony itself & acting upon receptors on OCT4 cells in an autocrine fashion was sufficient to yield pure PAX6 multipotent hNSCs (which could mature into electrophysiologically-active anterior dorsal cortical interneurons, which became caudalized with time, as well as other more specialized neural lineages, including glia, and motor and dopaminergic neurons). Inactivation of endogenous MDK (via neutralizing antibodies or shRNA-mediated knockdown) blocked neuralization. As suggested by the dataset and experimentally validated, MDK’s neural induction was mediated in part by PI3K/AKT/mTOR signaling, the inhibition of which disrupted neuralization and shifted hESC fate towards endoderm. Taken together, the (phospho)proteomic dataset and the confirmatory experiments above provide insights into a heretofore unrecognized mechanism critical to the emergence of neuroectoderm. TRACK-HD and TRACK-ON HD – yielding new insights into Huntington’s disease Sarah Tabrizi London, UK Huntington’s disease (HD) is a devastating autosomal dominantly inherited neurodegenerative disease for which there is currently no effective disease modifying therapy. The genetic predictability of HD provides an opportunity for early therapeutic intervention many years before overt symptom onset and at a time when reversal or prevention of neural dysfunction may still be possible. As HD is monogenetic, fully penetrant, and characterised by a long premanifest phase, it is emerging as a potential model for studying therapeutic intervention in other neurodegenerative conditions such as Alzheimer’s or Parkinson’s disease where no preclinical diagnostic tests exist. Understanding of HD pathogenesis is evolving, and there are a number of candidate therapeutics with potential diseasemodifying effects that are currently being tested. The most promising approaches will be briefly reviewed. Since 2008 TRACK-HD has chronicled the earliest stages of the neurodegenerative disease processes in premanifest and mild to moderately symptomatic individuals who carry the HD expansion mutation (Tabrizi et al., Lancet Neurology 2009, 2011). TRACK-HD was designed to observe natural disease progression in premanifest and early stage HD with the aim of understanding the preclinical and early phases of neurodegeneration, phenotypic correlates of neuronal dysfunction and to establish sensitive and specific clinical and biological markers of disease progression. In 2012, we reported longitudinal effect sizes for disease-progression in early stage HD over 24 months (Tabrizi et al., Lancet Neurology 2012). Both our 24 month and recently published 36 month time-point data will be presented (Tabrizi et al., Lancet Neurology 2013), including new insights into predictors of HD progression in both premanifest and early stage subjects, and a range of novel clinical measures that now show significant change in the premanifest group over this period. We have also identified baseline predictors of disease progression which may help enrichment for future disease-modifying clinical trials. We are now in a position to model progression in a range of functional, biochemical and imaging measures across the spectrum of disease. Finally I will summarise our ongoing research aiming to identify neural compensatory networks that may occur in the premanifest phase of neurodegeneration in HD, and dissection of the role of the innate immune system as a key modifier of disease progression. Growth factor gene therapy for Alzheimer’s disease: NGF and BDNF Mark H. Tuszynski La Jolla, CA 92903 Nervous system growth factors have extensive effects on neuronal function and survival. Nerve growth factor (NGF) prevents the death and stimulates the function of basal forebrain cholinergic neurons in correlational models of Alzheimer’s disease (AD), leading to its translation to Phase 1 and 2 human clinical trials. Separately, Brain-Derived Neurotrophic Factor (BDNF) influences the survival and function of entorhinal cortical and hippocampal neurons in several animal models of AD, including transgenic mutant APP-expressing mice; aged rats and lesioned rats; and aged and lesioned primates. Thus, BDNF therapy has the potential to specifically target memory deficits in AD. The beneficial effects of growth factors in AD models appear to occur independently of detectable alterations in beta amyloid load, providing therapeutic alternatives to AX-modifying therapies in AD. This lecture will provide an update on the current status and future directions of neurotrophic factor therapies that are in preclinical and clinical development. Adult stem cell-based therapy for ischemic stroke Catherine M. Verfaillie Leuven, Belgium 150 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Neural stem cells or neural stem/progenitor cells differentiated from pluripotent stem cells can generate neurons, astrocytes and oligodendrocytes, which could be used for regeneration of lost neural tissue following stroke, even if this has yet to be accomplished. Other cell populations are also being considered for therapy of stroke. We will describe the potential of (stem) cells from non-neural postnatal tissues, such as bone marrow, which themselves do not (or maybe in a very limited manner) generate neural progeny. However, these non-neuroectoderm derived cell populations may induce endogenous neurogenesis and angiogenesis, as well as have immunomodulatory properties that improve stroke outcomes. Preclinical animal studies (and pitfalls), early clinical studies, and possible mechanisms of action, as well as possible unforeseen complications will be discussed. From pluripotent stem cells to cortical circuits: perspectives for disease modelling and brain repair Pierre Vanderhaeghen Bruxelles, Belgium The cerebral cortex consists of several hundreds of different types of neurons, organized into specific cortical layers and areas, that display specific profiles of gene expression, morphology, excitability and connectivity. Embryonic stem (ES) and other pluripotent stem cells constitute a promising tool for the modelling and treatment of human neural diseases. Here we describe an intrinsic pathway by which pluripotent stem cells, whether of mouse or human origin, recapitulate in vitro the major milestones of cortical development, leading to the sequential generation of a diverse repertoire of pyramidal neurons that display most salient features of genuine cortical neurons. Interestingly, the mouse and human pathways of corticogenesis display many similarities but also striking differences that may be related to species-specific developmental programmes. When transplanted into the cerebral cortex of new-born mice, the ESC-derived cortical neurons develop specific patterns of axonal and dendritic projections corresponding to endogenous cortical projections in vivo. Following transplantation into lesioned adult mouse cortex, the grafted neurons also establish robust and specific long range projections and synapses corresponding to cortical circuits. These data shed new light on the mechanisms of neuronal specification, and constitutes an innovative tool to model human cortical development, evolution, and disease, in vitro and in vivo. In the long run, cortical neurons generated in vitro could be used also in the perspective of brain repair, for several diseases striking cortical neurons. Production of new striatal neurons in the healthy and diseased neonatal brain J. Wright and L.H. Thompson Melbourne, Australia Substantial advances have been made in the last decade on our understating of the basic physiology underlying neurogenesis in the postnatal mammalian brain. The bulk of the work in this area has been based on analysis of the adult brain. Relatively less is known about the capacity for neurogenesis in specific structures within the neonatal brain. Here we demonstrate that the production of medium spiny striatal projection neurons (MSNs) in the striatum extends into the early neonatal period under normal physiological conditions in the rat brain, with low but stable contributions of striatal interneurons during this time-period. Using retroviral labeling, we further show how MSNs born neonatally, correctly acquire their axonal targets in the globus pallidus and midbrain. These data provide evidence of latent MSN development in the neonatal striatum post-embryogenesis and as such, these new born cells provide promising targets for gene therapy to promote endogenous self-repair during neonatal striatal injury such as in cerebral palsy. Based on this, we are developing a novel hypoxia-independent neonatal stroke model in order to compare our results with striatal neurogenesis in a damaged brain which will give further insight into whether MSN production is enhanced or inhibited due to striatal damage. ABSTRACTS OF POSTER PRESENTATIONSS OF POSTER PRESENTATIONS Generating patient-derived Parkinsonian iPS cell lines, their differentiation to midbrain dopaminergic neurons and susceptibility to neurotoxic insults Peter A. Barbuti, Jennifer L. Badger, Lucy A. Crompton, Alan L. Whone, James B. Uney and Maeve A. Caldwell Bristol, UK Parkinson’s disease (PD) is neurodegenerative disease clinically characterised by the loss of A9 dopaminergic neurons in the substantia nigra pars compacta (SNc) of the midbrain. Using patient-derived stem cells we aim to model the disease by recapitulating the neurons lost in PD and investigate their susceptibility to neurotoxic insult. Dermal fibroblasts from idiopathic and genetic PD patients were transduced with lentiviruses to generate induced pluripotent stem (iPS) cell lines. We have developed a robust protocol for the generation of dopaminergic neurons of the SNc and have characterised these neurons from lines generated for the markers of SNc and their dopamine release. We have then subjected these neurons to neurotoxic insults to model PD in-vitro and are currently looking at ways to protect these de-novo neurons from toxicity and prevent their degeneration. Acknowledgements: This work was funded by Parkinson’s Disease UK. Abstracts 151 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Understanding rejection of allogeneic and xenogeneic dopaminergic cell transplants Ludivine S. Breger, Stephen B. Dunnett and Emma L. Lane Cardiff, Wales, UK A restorative approach for Parkinson’s disease (PD) is thetransplantation of foetal ventral mesencephalon (VM). Post-mortem studies of patients treated in previous clinical trialshave revealed the presence of infiltrated immune cells [1],which may result from a lack of immune compatibilitybetween the donor cells and the host. The impact this mayhave on the functional outcome of cell therapy for PD hasyet to be determined. To address this question, it isnecessary to establish a controlled model of rejection so thiswork compared the rejection of allograft and xenografttransplants in rat hosts. 6-OHDA unilaterally lesionedSprague Dawley received intrastriatal VM transplantderived from embryos of CD1 mice (xenograft) or Wistarrats (allograft). To establish the time course of rejection,xenografted animals received immunosuppressive treat-ment (cycloA) for different durations, while rejection ofthe allograft was obtained by peripheral injections of spleencells [2]. Rotational behavior in response to amphetaminewas used as a behavioural assessment of graft survivalalongside immunohistochemical analysis. The immuneresponse, determined by infiltration of leukocytes, appearswithin the second week of withdrawal of immunosuppres-sion, although no significant cell loss was observed beforethe 3rd week. Regarding the allograft model, the use ofdifferent time points for spleen cells injections induceddifferent degrees of rejection, from 23% to 96% grafted cellloss, 4 weeks post-graft, therefore creating a morecontrollable model than the xenograft model. References1 Kordower JH, Styren S, Clarke M, DeKosky ST, Olanow CW, Freeman TB.Fetal grafting for Parkinson’s disease: expression of immune markers in twopatients with functional fetal nigral implants. Cell Transplantation 1997;6:213–219.2 Soderstrom KE, Meredith G, Freeman TB, McGuire SO, Collier TJ,Sortwell CE, et al. The synaptic impact of the host immune response ina parkinsonian allograft rat model: Influence on graft-derived aberrantbehaviors. Neurobiology of Disease 2008; 32:229–242. A potential compensatory role for endogenous striataltyrosine hydroxylase-positive neurons in a non-humanprimate model of Parkinson’s diseaseAndrew N. Bubak, D. Eugene Redmond Jr,John D.Elsworth, Robert H. Roth, Timothy J. Collier,Kimberly B.Bjugstad, Barbara C. Blanchard andJohn R. Sladek JrDenver CO, New Haven CT and East Lansing MI, USA The neurotoxin MPTP (1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine) selectively destroys dopaminergic (DA)neurons in the nigrostriatal pathway while importantlysparing other DA systems, serving as a significant toolin Parkinson’s disease (PD) research. Extensive studies inour laboratories have shown reversal of MPTP induced PDsymptoms following striatal grafts of fetal ventral mesence-phalic DA neurons in the African Green monkey. Addition-ally, our and other studies have reported an upregulation ofendogenous tyrosine hydroxylase (TH) positive neurons inthe striatum following MPTP lesions (B140% increase).The aim of the current research is to investigate the fate ofthis endogenous population in MPTP treated monkeysfollowing fetal cell grafts. Preliminary results indicate areturn to control levels for the TH-positive cells followingsuccessful transplantation, while animals whose donor cellgrafts failed to contain DA neurons retained the elevatedlevels. If these cells represent a compensatory mechanismin an attempt to replenish DA in the striatum, then theability to influence this population could prove beneficial indeveloping new non-invasive therapeutic treatments.Current research investigating the origin of these cellsand their relationship to symptom severity is ongoing.Supported by an Academic Enrichment Grant from theUniversity of Colorado School of Medicine, The AxionResearch Foundation, and 5PO1NS044281. Human Induced Pluripotent Stem (iPS) Cells for CellReplacement Therapy in Huntington’s disease N. Choompoo, N.N. Vinh, C.M. Kelly and A.E. RosserCardiff, UK Huntington’s disease (HD) is a neurodegenerativedisease caused by a mutation in the huntingtin gene(HTT). The extended CAG repeat ultimately leads toloss of medium spiny neurons (MSNs) in the striatum ofthe HD brain. Cell replacement therapy using primaryhuman fetal tissue has shown ‘proof of principle’ as astrategy to treat this genetically inherited disease [1].However, alternative cell sources need to be identified toovercome the ethical and logistical issues that areassociated with using human fetuses. IPS cells were firstintroduced by Yamanaka in 2006 by direct reprogrammingof fibroblasts to ES-like cells by using four defined factors (Oct4, Sox2, Klf4, c-Myc) [2]. Here we attempt togenerate safety iPS cells by introduce reprogrammingfactors using piggyBac Transposon [3] transduction systemto human fetal fibroblasts and fetal neural stem cellswhich theoretically have the potential to be more readilyreprogrammed and are genetically unrelated to the host.The establishing iPS cell line were present the simila-rities to human embryonic stem (ES) cells in morphology, surface antigen, and proliferation. References1 Kelly MC, Dunnett SB, Rosser AE. Medium spiny neurons for transplantationin Huntington’s disease. Biochem Soc Trans 2009; 37:323–328.2 Takahashi K, Yamanaka S. Induction of pluripotent stem cells from adulthuman fibroblasts by defined factors. Cell 2007; 131:861–872.3 Woltjen K, et al. piggyBac transposition reprograms fibroblasts to inducedpluripotent stem cells. Nature 2009; 458:766–770.152 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Differential neuroprotective capacity of EndothelialProgenitor Cells-derived factors Stefano Di Santo, Stefanie Seiler and Hans R. WidmerBern, Switzerland Stem and progenitor cells release a wide array of trophicfactors, cytokines and extracellular matrix molecules. Thepresent study aimed at investigating whether endothelialprogenitor cells (EPC) may support neuronal functionsand survival by means of paracrine factors. EPC isolated from peripheral blood of healthy humandonors were cultured in hypoxic conditions to stimulatesecretion of growth factors. Primary cultures from fetal ratembryonic (E14) ganglionic eminence (GE) and ventralmesencephalon (VM) were treated with EPC derivedconditioned medium (EPC-CM). Incubation of cultureswith EPC-CM resulted in a significant increase in TH-ircell densities in VM as well as GABA-ir cell densities in theGE cultures, respectively. Interestingly, treatment ofcultures with EPC-CM resulted in a substantial increasein number of microglial cells. Notably, EPC-CM displayedneuroprotection against MPP+ toxicity in VM cultureswhereas it was not effective against 3NP toxicity. Theeffect of EPC-CM on TH-ir cells persisted upon treatmentwith Ara-C conducted in order to limit microglia expansion. Our findings identified EPC-CM as a powerful tool topromote viability and/or differentiation of neuronal cells.EPC-CM constituents might represent a new strategy tosupport neuronal repair. Supported by the Swiss National Science Foundation andthe Hanela Foundation. Characterisation of FoxP1 in the striatum A.E. Evans, M.V. Taylor and A.E. RosserCardiff, UK Medium Spiny Neurons (MSNs) constitute >90% of theprojection neurons in the striatum and are lost inHuntington’s disease (HD). A screen looking at gene expression changes in the developing striatum of mice (E12-E16) showed FoxP1 to be the most up-regulated gene overthis period. FoxP1 is also expressed in the adult striatum andhas the potential to be a marker of both developing andmature MSNs (Tamura et al., 2003). Understanding theregulation of striatal development, in particular, thedifferentiation of MSNs is the key to finding strategies to‘direct’ the differentiation of such cells from pluripotentstem cells. This in turn is crucial for generating genuinestriatal cells for cell therapy in conditions such as HD. To date the function of Foxp1 in the brain is unknown.Homozygous FOXP1 knock-outs (FoxP1 / ) in mice areembryonically lethal from BE15 due to cardiac defects(Wang et al., 2004). However, one can analyse the embryos before the onset of lethality. Using several techniques wehave attempted to understand what role FoxP1 has in MSNdifferentiation. Results show that there is a decrease in thenumber of DARPP-32 expressing neurons when FoxP1 isabsent. In addition, ongoing microarray analysis will high-light differences at a genomic level, between the FoxP1 / –nulls and WTs in the striatum at E14. Birth dating of dopamine neuron subtypes in ventralmesencephalon grafts in a rat model of Parkinson’sdisease M. Fjodorova, E.M. Torres and S.B. DunnettCardiff, UK Using co-expression of tyrosine hydroxylase (TH) withGIRK2 and Calbindin1 to identify A9 and A10 dopamineneuron subtypes, respectively, we have previously shownthat grafts derived from younger ventral midbrain donortissue are enriched with A9 neurons. It is unclearhowever, whether this is due to different survival levelsor differentiation of dopamine neuron precursors aftertransplantation. Here we used BrDU labelling to birth date rat embryonicday 12 (E12) and E14 dopamine neurons after transplanta-tion into the striatum. The results show that younger donorgrafts contained significantly more mitotic dopamineneuron precursors, which continued to proliferate in thehost striatum and two thirds of which were able to matureinto A9 neurons, a third differentiated into A10 neurons.These findings demonstrate that a higher yield offunctionally important A9 neurons in younger donor graftsis, at least in part, due to continued dopamine cell divisionafter transplantation. This has significant implications forthe generation and selection of dopamine precursors at theright developmental stage from stem cell based sources toimprove cell transplantation therapy in PD. Acknowledgements: This work was funded by the WellcomeTrust. Reference1 Thompson L, Barraud P, Andersson E, Kirik D, Björklund A. Identification ofdopaminergic neurons of nigral and ventral tegmental area subtypes in graftsof fetal ventral mesencephalon based on cell morphology, protein expression,and efferent projections. Journal of Neuroscience 2005; 25:6467–77. Vitamin D3 promotes dopamine neuron survival throughupregulation of GDNF Rowan P. Orme, Manminder Bhangal andRosemary A. FrickerKeele, UK Vitamins play a key role in brain development and neuronalsurvival. In previous work using an iTRAQ proteomics technique and immunohistochemistry we identified vita-min D3 receptor expression during peak neurogenesis ofdopamine neurons in the ventral midbrain. In this study we applied the active metabolite of vitaminD3, calcitriol, to cultures of embryonic day 12 ventralAbstracts 153 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. mesencephalon neurons to investigate its role in neuronaldifferentiation and survival. Calcitriol enhanced the totalnumber of tyrosine hydroxylase-positive dopamine neu-rons in a dose dependent manner, up-regulating GDNF,and proffering neuroprotection rather than generatingnew dopamine neurons. Blocking GDNF signallingreversed calcitriol’s positive effect on enhancing thenumber of dopamine neurons. We then treated E12 ventral mesencephalic cell suspen-sions with vitamin D3 either as calcitriol to the transplantsuspension or vitamin D3 delivered orally – prior totransplantation in 6-OHDA lesioned rats. Although allgrafts reversed amphetamine-induced rotation to a similardegree, vitamin D3-treated grafts gave earlier recovery inthe cylinder test compared with control transplants. Trans-plant histology to assess vitamin D3’s effects on dopamineneuron survival and graft-host innervation will be presented. Acknowledgements: This work was funded by Parkinson’s UKand Keele Medical School. The influence of nicotinamide on directed differentiationof neurons from embryonic stem cells in vitro Sı́le M. Griffin, Mark R. Pickard, Rowan P. Orme,Clive P. Hawkins and Rosemary A. FrickerKeele, Staffordshire, UK Factors controlling proliferation and differentiation arecrucial towards advancement of neural cell-based experi-mental neurodegenerative therapies. In this regard,nicotinamide has been shown to function in neural cellfate determination, enhance neuralization and influenceDNA repair and apoptosis. This study investigated whether nicotinamide could directthe differentiation of mouse embryonic stem cells (mESCs),cultured as monolayers, into neurons. 5mM and 10mMnicotinamide added at days 0-7 and 7-14 significantlyincreased b-III tubulin positive neuronal populations,concomitantly decreasing the total number of cells inculture measured by quantification of40,6-diamidino-2-phenylindole (DAPI) positive cells. Interestingly, nicotina-mide had a more significant effect on neuronal differentia-tion at day 0-7. Current work is focusing on elucidating themechanism mediating neural specification by nicotinamide,i.e. induction of cell-cycle exit and/or selective apoptosis innon-neural populations. Preliminary data indicates a reduc-tion in the proportion of proliferating cells in nicotinamide-treated cultures, suggesting that nicotinamide may enhance cell-cycle exit thereby promoting neuronal differentiation.We are currently investigating the effect of nicotinamide on the process of neural induction and whether it influencesneuronal subtypes generated in vitro. Future work will focus on evaluating the effect ofnicotinamide on the differentiation of midbrain dopamineneurons; towards a therapy for Parkinson’s disease.Brain endogenous Liver X Receptor ligands selectivelypromote midbrain neurogenesisSpyridon Theofilopoulos, Yuqin Wang,William J. Griffiths and Ernest ArenasStockholm, Sweden and Swansea UK Liver X receptors (Lxra and Lxrb) are ligand-dependentnuclear receptors critical for ventral midbrain neurogenesisin vivo. However, no endogenous midbrain Lxr ligand hasso far been identified. Here we employed LC-MS andfunctional assays to identify cholic acid as a novel Lxrligand. Moreover, 24(S),25-epoxycholesterol (24,25-EC)was found to be the most potent and abundant Lxr ligandin the developing mouse midbrain. Both Lxr ligandspromoted neural development, in an Lxr-dependentmanner, in zebrafish in vivo. Intriguingly, each ligandselectively regulated the development of distinct midbrainneuronal populations. While cholic acid increased survivaland neurogenesis of Brn3a+ Red Nucleus neurons, 24,25-EC promoted dopaminergic neurogenesis. These resultsidentify an entirely new class of highly selective and cell-type specific regulators of neurogenesis and neuronalsurvival. Moreover, 24,25-EC promoted dopaminergicdifferentiation of ES cells, suggesting that Lxr ligandsmay thus contribute to the development of cell replace-ment/regenerative therapies for Parkinson’s disease [1]. Acknowledgements: This work was funded by BBSRC andthe Swedish Research Council. Reference1 Theofilopoulos S,Wang Y, Kitambi SS, Sacchetti P, Sousa KM, Bodin K, et al.Brain endogenous liver X receptor ligands selectively promote midbrainneurogenesis. Nat Chem Biol 2013; 9:126–133. Safety study on cyclosporine A in epilepsy models Annelie Handreck, Eva M. Mall, Deborah A. Elger,Laura Gey and Manuela GernertHannover, Germany Neural transplantation of inhibitory cells into seizureinitiating or propagating brain regions is one promising approach to treat pharmacoresistant epilepsies. Depend-ing on the grafted cell type (e.g. xenotransplantation),immunosuppression is necessary to prevent graft rejec-tion, thereby enabling long-lasting anticonvulsant effectsof the graft. We therefore investigated the effects of daily treatment(15 days) with different preparations of the commonlyused immunosuppressive drug cyclosporine A (CsA) (pure substance or ready-to-use-drugSandimmun, No-vartis), doses (5 or 10mg/kg), and application routes (i.p. vs. s.c.) on acute and chronic seizure thresholds in rats.Additionally, CsA whole blood levels were analyzed andbehavioral tests were conducted to detect side effects. The data did not reveal acute and only subtle chronic effectsof immunosuppression with either pure CsA or Sandimmun154 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. on seizure thresholds. The resorption of i.p. applied CsAfrom Sandimmun exceeded the resorption from the pureCsA preparation. Unwanted side effects included transientgastrointestinal problems. Our data indicate that immuno-suppression with 10mg/kg Sandimmun i.p. rather than pureCsA is a safe and feasible option for use in neuraltransplantation experiments in epilepsy models. Acknowledgements: Supported by the DFG (Ge1103/7-2).AH is supported by the Prof. Dr. Peter and Jytte WolfFoundation for Epilepsy. Are transplant-induced improvements in cognitiveperformance in the rat lesion model of Huntington’sDisease dependent on frontal-striatal circuitreconnection? D.J. Harrison, S.P. Brooks and S.B. DunnettCardiff, UK Frontal-type executive function, demonstrated in rats usinga delayed alternation (DA) task, is disrupted whencontinuity of both cortico-striatal loops of the associatedneural circuitry are interrupted. Bilateral grafts of embryo-nic whole ganglionic eminences (WGEs) restore function inthe DA task in bilateral striatally lesioned rats, withtransplanted tissue integrating with the host brain.However, it has not been definitively shown that improve-ment in cognitive performance is due to physicalreconstruction of neural circuitry or if grafted tissue simplyexhibits other beneficial influences on the host brain, forexample by releasing donor derived growth factors. There-fore, in the present study, we aim to determine the role ofcircuit reconstruction in the attenuation of cognitivedeficits. This is to be assessed by training rats on theoperant DA task and evaluating performance followingbilateral striatal lesions, unilateral striatal transplantation ofWGE, and ipsilateral or contralateral surgical disconnectionof the prefrontal cortico-striatal circuitry. We hypothesise that unilateral grafts should provideeffective alleviation of the bilateral striatal lesion deficit,and that contralateral knife cuts will preserve prefrontalcortical afferents to the grafted striatum thus maintainingcognitive performance, whereas ipsilateral disconnectionof the graft will reinstate the deficit. The data presentedrepresents the current status of the study. This work is supported by the MRC. Optogenetic manipulation of hESCs and in vivomeasurement of dopamine release Andreas Heuer, Shane Grealish, Agnete Kirkeby,Martin Lundblad and Malin ParmarLund, Sweden Human embryonic stem cells (hESCs) are a possible alternative to primary fetal tissue for the transplantation ofdopamine neurons into patients with Parkinson’s disease(PD). We aim to investigate the contribution of engraftedhESC-derived mesencephalic dopamine (mesDA) neuronson functional improvements in a rat model of PD. In this study we use Channelrhodpsin (ChR2) allowingus to manipulate cell activity and release DA with lightpulse stimulation, at a high temporal resolution. Bytransducing hESCs with an LV-vector to express ChR2under the synapsin-1 promoter (Syn), transplanted cellswill express the construct in neurons. Here we engrafted one group of 6-hydroxydopaminelesioned rats with H9-hESC-Syn-ChR2 cells that werepatterned towards a mesDA phenotype whilst having alesioned group serving as control. We will assess cellsurvival and dopamine release of transplanted cells viaamphetamine-induced rotations as well as using in vivoamperometry in conjunction with optogenetic stimulation, in order to manipulate neural activity. At the conferencewe will present the first findings of this study. It is hopedthe results from this study will provide insight into theneurochemical events that underpin graft-mediated DA-release/functional recovery in rat models of PD. Influence of transcranial direct current stimulation onthe survival, migration and integration of dopaminergiccell transplants in a rat model of Parkinsońs disease:a histological and behavioral analysis B. Fritsch, N. Hoffmann, L. Furlanetti, J. Garcı́a,M.D. Döbrössy and C. WinklerFreiburg, Germany Introduction: Parkinsońs disease (PD) is a challengingdisease, which leads to progressive and disabling dete-rioration of motor and cognitive skills. Transcranial directcurrent stimulation (tDCS) is a non-invasive techniquethat presents modulatory effects on cognitive functionsand motor behavior. Regenerative approaches are underresearch to restitute dopaminergic neurotransmission andoffer a more extensive and long lasting effect. In thisstudy we analyzed the effect of tDCS on the survival, migration and integration of dopaminergic cell transplants. Methods: Rats received unilateral dopamine-denervatinglesions using 6-hydroxydopamine. Afterwards, they wererandomly assigned into three groups (Anodal-Stim, Cath-odal-Stim and Sham-Stim) according to values in amphe-tamine-induced rotation. All groups were transplanted withE14 GFP ventral mesencephalic (VM) cells into theipsilateral striatum. tDCS was started right after transplantation and performed for 20min daily, during 2 weeks.Amphetamine-induced rotation and cylinder test were performed pre transplantation and 5 weeks after transplan-tation, respectively. Rats were perfused 5 weeks aftertransplantation and histological analysis was performed. Results: Survival of transplanted VM cells in the striatumwas increased by approximately 50% in the anodallyAbstracts 155 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. stimulated group as compared to sham and cathodal groups,and optical fiber density was increased in the anodal tDCS-group. All groups improved their scores in amphetamine-induced rotation, but there were not improvements in thecylinder test at this early time-point after grafting. Conclusion: Transcranial direct current stimulation im-proved graft survival and fiber outgrowth of intrastriataldopamine grafts in a rat PD model. Further studies areongoing to confirm and extend these findings. Trefoil Factor 1 in the nigrostriatal system of6-hydroxydopamine-lesioned ratsPia Jensen, Michel Heimberg, Angelique D. Ducray,Hans R. Widmer and Morten MeyerOdense, Denmark and Bern, Switzerland Trefoil factor 1 (TFF1) belongs to a family of secretedpeptides mainly expressed in the gastrointestinal tract.TFF1 has also been suggested as a neuropeptide, but notmuch is known about its expression and function in theCNS. We investigated the expression of TFF1 in control and6-hydroxydopamine (6-OHDA)-lesioned rats. In theunlesioned ventral mesencephalon, TFF1-immuno-reactive (-ir) cells were mainly found in substantia nigrapars compacta (SNc) and in the ventral tegmental area.While around 90% of the TFF1-ir cells in SNc co-expressed tyrosine hydroxylase (TH), only a subpopula-tion of TH-ir neurons expressed TFF1. Some TFF1-ircells in SNc co-expressed calbindin or calretinin, andnearly all were NeuN-ir while there was no co-localizationwith the astroglial marker GFAP. A few TFF1-ir cells werealso found in the striatum, and their numbers signifi-cantly increased after 6-OHDA lesion. Intrastriatal injection of Fluorogold resulted in retrogradelabeling of several TFF1-ir cells in the SNc showing thatthese cells were projection neurons. This was alsoreflected by unilateral loss of TFF1-ir cells in the SNc of 6-OHDA-lesioned rats. Our findings demonstrate that distinct subpopulationsof midbrain dopaminergic neurons express TFF1 and that this expression pattern is altered in a rat modelof Parkinson’s disease. Comparison of L-Dopa response patterns of differentstages of MSA-P in a double lesion rat modelChristine Kaindlstorfer, Joanna Garcı́a,NadiaStefanova, Werner Poewe, Christian Winkler,Mate Döbrössy and Gregor WenningInnsbruck, Austria and Freiburg, Germany The objective of this study was to identify the striatalsub-region and QA dosage replicating L-Dopa failure in apartial double lesion rat model of MSA-P. Threeexperimental groups were investigated: (1) PD groupreceiving 6-OHDA lesion, (2) MSA-P mild (3) MSA-Psevere group receiving 6-OHDA and QA lesion. Following6-OHDA lesion all groups showed significant behavioraldeficits in cylinder and stepping test, without furtherdeterioration after QA lesion except MSA-severe groupwith significant impairment in stepping test. Ampheta-mine induced rotation resulted in ipsilateral rotation ofPD and MSA groups, whereas apomorphine inducedcontralateral rotation in PD and ipsilateral rotation inMSA groups. All groups revealed a significant L-Doparesponse in cylinder and stepping test after 6-OHDAlesion that was significantly reduced in both MSA groupsfollowing the subsequent QA lesion, with a trend of MSA-P severe group being most resistant to L-Dopa. Inconclusion, the current study describes the unique motorbehavior and L-Dopa response patterns in association tothe location and the dosage of the QA lesion of doublelesioned animals compared to single 6-OHDA lesionedanimals. Both QA lesion approaches in MSA-mild andMSA-severe groups led to a reduction of L-Doparesponsiveness, whereas the higher dose of QA showedthe greatest reduction of such. This refined model ofMSA-P replicating L-Dopa failure is highly suitable forfuture grafting studies with the rational of restoring L-Dopa responsiveness. Cell intrinsic and extrinsic factors contribute to enhanceneural circuit reconstruction following transplantationin Parkinsonian mice Jessica Kauhausen, Lachlan Thompson and Clare ParishMelbourne, Australia Homotopic grafting into the SNpc is capable of restoringthe nigrostriatal pathway and forming appropriate con-nections with the striatum, but the extent of striatalreinnervation remains substantially less than can beachieved with ectopic placement. The aim of thisinvestigation was to determine what effect donor ageand virally delivered glial derived neurotrophic factor(GDNF) over-expression had on the survival, growth andintegration of homotopic grafts placed into 6OHDAmouse model of Parkinson’s disease. Younger donor tissue(E10) showed to be capable of generating larger graftswith more axonal growth compared to grafts generated from the more conventional E12 donor VM. With theaddition of GDNF, fiber growth and striatal innervationwas further enhanced in both older and younger donortissue. Immunohistochemical staining revealed thatyounger donor grafts treated with GDNF resulted in anincreased number and proportion of A9 DA neurons. Behavioural testing confirmed that younger donor graftsprovided a significant improvement in motor behaviorwith GDNF overexpression only affecting E12 donortissue. These findings could have significant implications156 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. for the future development of cell replacement therapyfor the treatment of Parkinson’s disease. Neonatal immune-tolerance in mice does not preventxenograft rejection: a comprehensive analysis usingin vivo luciferase trackingVirginia B. Mattis, Dustin R. Wakeman, Colton Tom,Hemraj B.Dodiya, Jack Reidling, Andrew Tran,Ksenija Bernau, Loren Ornelas, Anais Sahabian,DhruvSareen, Leslie M. Thompson,Jeffrey H. Kordower and Clive N. SvendsenLos Angeles, CA, Chicago IL, Irvine CA and Madison WI, USA While in vitro iPSC models are proving extremelyinformative, an in vivo ‘‘humanized’’ chimeric animalmodel of disease via transplantation of diseased humaniPSC-derived cells may prove ideal for therapeuticscreening and mechanistic discovery. One of the majorchallenges for the field is appropriate immune suppres-sion in these xenograft models as it is often ineffective orcost-prohibitive for long term studies, and has also beenshown to ameliorate neurological diseases (Rosenstocket al. Neurochem Int. 2011), complicating experimentalresults. With the promise of transplantation for neurode-generative diseases, there also is the need for non-invasivein vivo tracking. In the current collaborative study we used a range oftechniques and cells to establish tolerance of the neonataland adult rodent brain to neural xenografts. We show thatin contrast to rats, mouse neonates are sensitive to humanneural xenografts (from iPSCs, ESCs or fetal tissues).In three mouse strains, prior sensitization had no effecton the severe rejection of the cells. Luciferase imagingwas shown to be a powerful predictor of graft survival inthe striatum. Together these studies show that neonatalmice reject human cells, and that immune tolerancetechniques are not sufficient to prevent rejection inadult mice. Human fetal dopaminergic precursor celltransplantation: LIF-nanotherapy to promotegraft survival.S.M. Metcalfe, J.W. Zhao, P. Tyers, X. He, T.M. Fahmyand RA BarkerCambridge, UK and New Haven CT, USA LIF (Leukaemia Inhibitory Factor) is both a neuropoieticand a tolerogenic cytokine. Thus LIF represents a promising candidate for treating neural cell grafts topromote their survival whilst simultaneously suppressing any immune reactivity against the graft. Since in vivo use of LIF is hindered by its rapid degradationand excretion, we have prepared a nano-particulateformulation of LIF intermixed with PGLA, a biocompa-tible, biodegradable polymer similar to that used in solublesutures. These "LIF-nano" are decorated with avidinallowing attachment of biotinylated anti-Thy1 to bind theparticles to neural precursor cells prior to grafting. LIF also plays a role in supporting neural stem andprecursor cells, and may provide dual therapeutic benefitsfor cell transplantation in Parkinson’s Disease (PD). TheTransEuro Project (http://www.transeuro.org.uk) is currentlyevaluating grafts of human fetal ventral mesencephalon(hfVM, rich in precursor dopaminergic cells) in treatmentof PD. Working in parallel, we have shown that, ex vivo,LIF-nano-treated hfVM yield over 3-fold more dopami-nergic cells compared to empty-nano controls. Similarly,in vivo, intra-striatal hfVM grafts in nude rats show majorbeneficial effects of LIF-nano therapy, both in graftsurvival and in dopaminergic cell maturation. This has major implications for clinical transplantation, reducing numbers of cells required for therapeutic effect. This work is supported by an NIHR i4i Grant. Simultaneous in vivo monitoring of transplanted cellsand biomaterials by magnetic resonance imagingWen Ling, Francesca Nicholls, Daniela delli Castelli,Christopher J.Medberry, Tao Jin, Seongi Kim,Stephen F.Badylak, Silvio Aime and Michel ModoPittsburgh, USA, London, UK and Turin, Italy Non-invasive in vivo monitoring of regenerative medicineapproaches for tissue and organ reconstruction will be animportant technological development to ensure the safetyand efficacy of such strategies. However, the involvement ofmultiple types of cells as well as biomaterials complicatesin vivo monitoring that is dependent on a single contrastmechanism. Herein we explored the use of chemicalexchange saturation transfer (CEST) to specifically visualizedifferent elements (multiple cell types and biomaterials)used for an in situ regenerative medicine approach for the treatment of the stroke-damaged brain. Specifically,neural stem cells and endothelial cells were labelledusing paramagnetic CEST (PARA-CEST) agents, whereasan extracellular matrix (ECM)-derived bioscaffold wasdetected using diamagnetic CEST (DIA-CEST). Intracel-lular incorporation of PARA-CESTagents (Eu-HPDO3A andYb-HPDO3A) was titrated to exert minimal biologicaleffects on the cells while ensuring sufficient levels wereachieved to afford detection in vitro and in vivo. Z spectrawere acquired for labelled cells as well as ECM bioscaffoldprior to ex vivo and in vivo imaging in a rat model of stroke.Although significant challenges remain, these results provideproof-of-principle that non-invasive imaging of tissue engineering in the brain is feasible. Acknowledgements: Funded by the European Union(201842ENCITE), the Department of Health ofPennsylvania (4100061184), and the Departmentof Radiology, University of Pittsburgh, USA.Abstracts 157 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. EGFP-Flag transfected rat progenitor cells displayelectrophysiological properties of integrated functionaldopaminergic (DA) neurons similar to geneticallylabelled DA-neurons in vitro and intrastriatallyxenografted DA-neurons in a rat model of Parkinson’sdiseaseMeltem Özer, Regina Rumpel, Martin Fischer, MalteDonert, Alexander Klein, Maike Wesemann, AndreasRatzka, Anna Effenberg, Christoph Fahlke andClaudia GrotheHannover and Jülich, Germany Whole cell patch clamp studies of intrastriatally graftedEGFP-Flag transfected ventral mesencephalic (VM) pro-genitor cells in brain slices displayed 2 types of neuronsshowing dopaminergic (Da)-like properties marked by thehyperpolarisation activated anomalous rectification. Type-1neurons display hyperpolarisation activated rectificationfollowed by delayed, single action potentials, and pace-maker activity. Type-2 neurons attract attention by distinctspontaneous bursting and bursting after hyperpolarisation.To clarify whether one or both Da-like neurons are actuallydopaminergic, fetal VM progenitor cells of a transgenic TH-GFP mouse were used to visualise Da neurons, aftergrafting in a 6-OHDA-rat model, directly due to expressionof fluorescent labelled tyrosine hydroxylase. These TH-GFP mice-derived Da-neurons we previously electrophy-siologically characterized in vitro. Since 6-OHDA lesion induces reduction in the ability touse the contralateral paw, an evaluation of motor skills bya staircase test can monitor functional integration of thegrafts. This is an alternative behavioural test to drug-induced rotation, which might influence the electro-physiological properties in situ. Overall, patch clamp analysis of genetically labelledDa-neurons together with behavioural studies facilitatethe direct investigation of functional properties of Daneurons in vitro and after intrastriatal grafting. Histopathological effects of varying the impacttrajectoriesin experimental model of TBI Mibel M. Pabón, Naoki Tajiri, Kazutaka Shinozuka,Sandra Acosta, Hiroto Ishikawa,Diana Hernandez-Ontiveroz, Julie Vasconcellos,Travis Dailey, Christopher Metcalf, Meaghan Staples,Cyrus Tamboli, Yuji Kaneko and Cesar V. BorlonganTampa FL, USA About 1.7 million people sustain TBI annually leading tosignificant disability and death around the world. Most ofthe TBI animal models focus on a single 90-degreetrajectory affecting a specific brain area; but TBI patients do not always display injury to the same brain regions.We examined histopathological effects at 3 days aftercontrolled cortical impact (CCI) model in adult Sprague-Dawley rats. We hypothesized that different impacttrajectories would produce varying levels of brain damage.CCI manipulations included: 1. conventional TBI target-ing frontal cortex, 2. farthest right angle (FRA) and3. closest right angle (CRA) both also targeting frontalcortex, 4. olfactory bulb (OB), and 5. cerebellar (CB)injury. Data showed typical cell loss in M1 cortical regionin the conventional TBI, FRA, and CRA groups usingH&E; staining and Cavalieri method. No significant celldeath was detected in M1 region of animals that receivedOB or CB injury. Comparable CA3 hippocampal cell lossin both hemispheres was noted across TBI groups.Increased expression of activated microglial marker OX-6 in M1 region accompanied conventional TBI, FRA, andCRA suggesting a critical inflammatory role. Theseresults provide a closer clinical approximation of varyingTBI histopathological outcomes following different im-pact trajectories. Acknowledgements: United States Department of Defense. Characterizing GDNF regulation and its impact in the6-OHDA rodent model Luis Quintino, Erika Elgstrand-Wettergren,Giuseppe Manfré, Christina Isaksson andCecilia LundbergLund, Sweden Destabilizing domains (DD) regulate gene expression bytargeting specific proteins fused to DD for destruction.More importantly, DD-regulated transgene expression canbe stabilized using small molecules such as trimethoprim. Lentiviral vectors were used to deliver regulated glialcell-derived neurotrophic factor (GDNF-DD) to thestriatum of rats. The expression of GDNF-DD was thenturned ON using TMP in the drinking water. Threeweeks after the expression was turned ON, the animalswere lesioned with 6-OHDA. Initial studies showed that GDNF-DD was able toprovide neuroprotection of substantia nigra neurons tolevels comparable to wild-type GDNF. Histologicalanalysis for GDNF activity using phoshorylated ribosomalprotein S6 (pS6) showed a significant increase in thenumber of surviving pS6 positive nigral cells of 140% inthe GDNF group and 77% in the GDNF-DD group where expression was turned ON. When the expression ofGDNF-DD was turned OFF, only 30% of pS6 positivecells remained in the lesioned substantia nigra. Thisdecrease was comparable to control groups. To determine if GDNF-DD expression could ameliorateor stop an ongoing degeneration or a fully lesioned brain, the expression of GDNF-DD was turned ON right after6-OHDA lesion or 4 weeks after the animals werelesioned with 6-OHDA. Amphetamine-induced rotations were performed beforelesion, 4 weeks after lesion and 9 weeks after lesion. In158 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. both groups where GDNF-DD expression was turned ONafter immediately after lesion and 4 weeks after lesionthere was a maintenance in the level of rotations. Thiswas in contrast with control groups, where there was anincrease of rotations through time, which reflected aprogressive degeneration of SNpc. Histological analysis using pS6 also indicated a significantincrease in the number of pS6 positive cells in bothgroups. The results indicate that the DD system is apromissing tool for regulating GDNF in vivo. Neonatal desensitisation to human embryonic tissuecan be induced using a range of tissue types Victoria H. Roberton, Claire M. Kelly and Anne E. RosserCardiff, Wales, UK Prior to clinical application; safety and efficacy of potentialhuman donor cells for transplantation in neurodegenera-tive disease must first be demonstrated in animal models.Neural xenografts are rejected by the rodent immunesystem within three weeks of implantation. Althoughimmunosuppressant drugs are mostly effective short-term,toxic side-effects lead to termination of experimentsbefore human cell differentiation. Immunodeficient hostscannot withstand rigorous functional testing, renderingcurrent methods inadequate for full preclinical assess-ment. We have developed a novel method in which hostanimals are ‘‘desensitised’’ to xenogeneic donor cells viaan injection during the neonatal period, promotingacceptance of human neural xenografts in adulthood. Successful desensitisation has been demonstrated in ratsusing neonatal injections of human neural tissue. Desensi-tising with non-neural cells would release more valuableneural tissue for transplantation and would also inform onthe immunological mechanisms underlying neonatal desen-sitisation. We compared survival of human transplants in therat striatum in hosts desensitised neonatally with differenthuman foetal tissues. Results suggest it may not be necessary for desensitising cell suspensions and trans-planted cells to match, but that some tissue types may bemore effective for desensitisation to xenogeneic tissue. Acknowledgements: Work funded by the Wellcome Trust. Controlled release of neurotransmitters frombiopolymer matrices influence neural stem cellproliferation and distributionN. Romanyuk, M. Vetrik, K. Karová, P. Jendelová,M.Hruby, J. Price and E. SykováPrague, Czech Republic and London, UK Spinal cord injury (SCI) results in cavity formation andthe loss of nervous tissue. Possible treatment cancombine stem cells with matrix support and drug releasein order to optimise the regeneration of lesioned tissue.We have utilized a heterogenic hydrogel system whichreleases cationic promoters that influence neural growth.The heterogenic ion exchanger Dowex 50WX8 wasscattered in porous hydrogels based on 2-hydroxyethyl-methacrylate or 2-hydroxypropylmethacrylamide. Neuro-transmitters and/or their analogs (carbachol, serotonin,L-dopamine and tryptamine) were bound to the ionexchanger. The release of carbachol and tryptamineincreased cell proliferation by 20–30% compared toneural stem cells cultured in the presence of a controlgel. Dopamine and serotonin significantly decreased cellproliferation. The growth and distribution of the cells inall hydrogels were analyzed immunohistochemically after 2,7, 14, 21 and 28 days. We demonstrated that the cells wereunable to attach and grow on the gel surface in thepresence of dopamine, whereas tryptamine and carbacholsupported both cell attachment and growth during 28 days. Our in vitro results demonstrate that the most suitablecandidate for further in vivo experiments examining thecombined treatment of SCI is a heterogenic hydrogelsystem releasing carbachol. Acknowledgements: P108/10/1560, 13-00939S. Transplantation of fetal ventral mesencephalicprogenitor cells overexpressing high molecular weightFGF-2 isoforms in 6-OHDA lesioned rats Regina Rumpel, Alexander Klein, Andreas Ratzka,Meltem Özer, Maike Wesemann and Claudia GrotheHannover, Germany FGF-2 is one of the most potent neurotrophic factorsto promote survival of dopaminergic (DA) neurons.It is expressed in different isoforms representingdifferent translation products from a single mRNA.For this study we selected the high molecular weight(HMW) isoforms (21/23 kDa) since we could show thatHMW FGF-2 transfected ventral mesencephalic (VM)cell preparations contained up to 3 times more DAneurons. After expansion and differentiation in vitro, HMW FGF-2 overexpressing embryonic day-12 VM cellswere transplanted unilaterally into 6-OHDA lesionedrats. Goal of this ongoing study is to analyze the effects ofHMW FGF-2 produced by the transfected VM cells astheir ‘‘own’’ neurotrophic factor. The experimental designcompares three grafted groups (HMW FGF-2 transfected,empty control vector transfected and non-transfected cells) with a lesioned control group. In addition, twodifferent transplantation paradigms with a differentamount of grafted cells are under evaluation. Animalsare analyzed for behavioural performance for up to 12weeks post transplantation and immunohistochemicalmeasurements of DA neuron numbers and striatal reinnervation are studied. Characterizing these grafts willaim to advance understanding of the influence ofAbstracts 159 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. transfected neurotrophic factors on survival and differ-entiation of DA cells. Antagonizing Nogo-receptor 1 promotes the numberof cultured dopaminergic neurons and elongatestheir neurites Stefanie Seiler, Dario Pollini, Stefano Di Santo andHans R. WidmerBern, Switzerland The myelin associated protein Nogo-A and its receptorNgR1 are among others the most potent growthinhibitors of the adult CNS. Nogo-A is mostly expressedon the surface of oligodendrocytes, but is also found inneurons of the adult and developing CNS. This suggeststhat Nogo-A serves additional functions in the brain. Inthe present study, we investigated the effects ofantagonizing NgR1 on dopaminergic neurons. For thatpurpose ventral mesencephalic (VM) cultures from E14rat embryos were grown in absence or presence of theNgR1 antagonist NEP1-40 for one week. Treatment withNEP1-40 significantly increased cell density of tyrosinehydroxylase (TH)-immunoreactive neurons. Morphologi-cal analysis of TH-positive neurons disclosed longerneurites and higher numbers of primary neurites incultures incubated with NEP1-40, while soma size wasnot changed. Moreover, organotypic VM culturesdisplayed significantly bigger volume and higherTH-positive cell numbers after NEP1-40 treatment.Similarly, Western Blot analyzes showed increased ex-pression levels of TH after NEP1-40 administration. In sum, our findings demonstrate that the intervention ofNogo-A signalling by antagonizing NgR1 modulatesdopaminergic neuron properties in the developing mid-brain. These observations might have substantial impactin the context of Parkinson’s disease. Supported by SNFand the Swiss Parkinson Foundation. The anti-dyskinetic effect of dopamine receptorblockade is enhanced in parkinsonian rats followingdopamine neuron transplantationEunju Shin, Carlo Lisci, Elisabetta Tronci,CaminoFidalgo, Roberto Stancampiano,Anders Björklund and Manolo CartaLund, Sweden and Monserrato, Italy We have previously shown that low doses of buspirone(5HT1A agonist and D2 receptor antagonist) and eticlo-pride (D2 receptor antagonist) can suppress graft-induceddyskinesia (GID) but are ineffective against L-DOPA-induced dyskinesia (LID) in a rat model of Parkinson’sdisease. However, while the effect against GID wasevaluated in grafted rats, the effect against LID wasinvestigated only in non-grafted animals. Therefore, thepresent study was performed to investigate whether thegraft might enhance the responsiveness of host striatumto these anti-dyskinetic drugs. If so, the same compoundsshould reduce LID only in grafted rats but not in non-grafted controls. Low doses of eticlopride (0.015mg/kg),SCH23390 (0.1mg/kg, D1 receptor antagonist), andbuspirone (0.3mg/kg) substantially reduced GID. Asreported before, anti-GID effect of buspirone was notprevented by a selective 5-HT1A antagonist, suggestingthat its effect is independent from 5-HT1A receptoragonism. Interestingly, the three compounds also po-tently reduced LID (induced by both 6 and 12mg/kg L-DOPA) in grafted rats but were ineffective in non-grafteddyskinetic controls. Taken together, these data demon-strate that the dopamine cell grafts strikingly exacerbatethe anti-dyskinetic effect of D1 and D2 antagonistsagainst both GID and LID, and suggest that the anti-GID effect of buspirone in patients may be due toblockade of D2 receptors. Proof-of-concept: The use of human induced pluripotentstem cells (hiPSCs) to uncover a novel developmentally-based target of therapeutic lithium in bipolardisease (BPD)B.T.D. Tobe, A.M. Crain, A.M. Winquist, M. Sidor,B.Calabrese, M. Brande, C. Duerr, M. McCarthy,C.McClung, S. Halpain, I. Singec and E.Y. SnyderLa Jolla CA and Pittsburgh PA, USA Neuropsychiatric disorders are difficult to model not onlybecause their non-specific multigene pattern, but alsobecause of the subjectivity by which they are oftendiagnosed. BPD, a highly-lethal illness, is unique in that50% of patients respond to lithium. Indeed, lithium-responsiveness is often pathognomonic. Critically,lithium’s mechanism-of-action is unknown; however,were its targets to be identified, lithium could providea molecular handle for discerning underlying mechanisms& deriving better treatments (lithium has unacceptableside-effects). We generated hiPSCs from lithium-respon-sive BPD patients from which neurons were generated.By performing differential proteomic analysis of ‘‘BPDneurons’’ exposed or not-exposed to lithium, we dis-covered a heretofore unanticipated lithium target,‘‘CRMP2’’, which plays a developmental role in neuriteextension, cell migration, & channel activity by virtue ofits association with tau & tubulin. We determined thatGSK3b & IPP2A regulate CRMP2’s interaction withcytoskeleton; excessive CRMP2 phosphorylation causes neurite retraction. Li robustly reduces CRMP2 phospho-rylation, validated in vivo in mouse hippocampus. Studiesare ongoing supporting the prediction that CRMP2mutation alters lithium responsiveness in mouse modelsof mania. Human material confirms an associationbetween CRMP2 & BPD. We offer a strategy for merging hiPSC technology with proteomics to develop moreeffective neurotherapeutic drugs.160 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Influx of blood monocytes to the brain is enhanced byantioxidants A. Rehnmark, P. Lopes, E. Faegermann, G. Orädd,A. Virel and I. StrömbergUmeå, Sweden Grafting in Parkinson’s disease involves implantation of fetaltissue into the brain where an on-going robust neuroin-flammation is present. The impact of the inflammation isstill elusive, and therefore this study was focused on theneuroinflammatory process in the striatal 6-hydroxydopamine(6-OHDA) model of Parkinson’s disease and the effects ofantioxidant treatment. It is well known that the striatalneurotoxic injection causes neuroinflammation, however, theorigin of the reactive microglia is not known, and suggestionshave been made that some of the inflammatory cells arebone-marrow-derived. This has been studied utilizingradiation of the bone-marrow cells and implantation oftagged cells, however, this technique opens the blood-brainbarrier. In this study, superparamagnetic iron oxide (SPIO)particles were combined with MRI. The SPIO particles werei.v. injected 24h prior to the 6-OHDA lesion was performed.The rats were scanned for T2*-weighted images at 1 weekpost-lesion using a 9.4T Bruker BioSpec. T2*-weightedimages are sensitive to iron, and the images becomehypointense. The results revealed no significant differencebetween animals that received a striatal lesion with orwithout SPIO particles. Giving the animals antioxidants inthe form of bilberry-enriched diet resulted in a significantstriatal hypointensity compared to controls. The SPIO-particles were tagged with a fluerescent marker andimmunohistochemical evaluations demonstrated that theparticles were found in the lesioned striatum of antiox-idant-fed rats. In animals given bilberry-enriched diet,striatal dopamine regeneration was found at later timepoints. Taken together, antioxidants promote influx ofbone-marrow-derived monocytes to the injured area, andresults in dopamine regeneration. A microRNA profile of reduced mir-34b/c and increasedmir-592 in adult epileptic patient-derived brain cellsreveals potent disease biomarker and screening tool fortransplantable stem cells Naoki Tajiri, Hiroto Ishikawa, Kazutaka Shinozuka,Travis Dailey, Robert Sullivan, Yuji Kaneko,Teresita Malapira, Carmelina Gemma, Fernando Vale andCesar V. BorlonganTampa, Florida, USA This study reports the utility of microRNA profiling as abiomarker and therapy development for temporal lobeepilepsy (TLE). Expression profiling of microRNAs (miRNAs), confirmed by qRT-PCR, revealed that miR-34b and miR-34c were significantly up-regulated in thehippocampus and amygdala compared to the neocortex,with miR-34b/c expression highest in the amygdala. Incontrast, levels of miR-592 were significantly down-regulated in the hippocampus and amygdala compared tothe neocortex. Immunocytochemical analyses providedinsights into the functional role of these miRNAs,demonstrating decreased cell proliferation and differentia-tion in the hippocampus and amygdala compared to theneocortex of TLE patients. Stem cell grafts from theneocortex (i.e., reduced miR-34b/c but elevated miR-592expression) survived in the amygdala, migrated to thelesioned hippocampus, and rescued hippocampal cell lossfrom the kainic acid-induced epilepsy in adult rats. ELISArevealed brain-derived neurotrophic factor (BDNF), glialcell line-derived neurotrophic factor (GDNF), and nervegrowth factor (NGF) were significantly down-regulated inthe human hippocampus and amygdala compared to theneocortex, while basic fibroblast growth factor (bFGF) wassignificantly up-regulated in the hippocampus and amyg-dala compared to the neocortex. These results support theuse of microRNA profiling as a biomarker and a screeningtool for stem cell-based therapies targeting epilepsy. Generation of cortical interneuron subtypes fromhuman pluripotent stem cells Claudia Tamburini, Charles Arber and Meng LiCardiff, UK Cortical interneurons are heterogeneous local inhibitoryneurons playing an essential role in maintaining abalanced activity in the cortex. Dysfunctions of thesecells have been associated with several neurological andpsychiatric disorders, such as schizophrenia, autism andepilepsy. Patient induced pluripotent stem cell (iPSC)-derived cortical neurons offer great promise for modellingthese diseases and serve as a platform for drug discovery.Furthermore, transplantation of stem cell-derived corticalinterneurons may be developed as a therapy for epilepsy. However, comparing to some other clinically relevantneuronal cell types (e.g. dopamine neurons), our ability todirect cortical interneuron differentiation from humanPSCs remains limited. We showed recently that Activinpromotes neuronal differentiation by inhibiting themitogenic Sonic Hedgehog pathway while enhancingthe pro-neurogenic retinoic acid signalling. In addition,Activin promotes the acquisition of a Calretinin inter-neuron fate by providing a caudal ganglionic eminenceidentity in PSC-derived neural progenitors. Extending these findings, our current work employs differentcombinations of morphogens to induce MGE-like pro-genitors, which subsequently give rise to Parvalbumin andSomatostatin expressing interneuron subtypes. Acknowledgements: This work was funded by the UKMedical Research Council. Recovery neurobiology of spinal cord injury: mechanismsgleaned from peripheral neurotization studies Yang D. Teng, Denise Konya, Dou Yu, Jamie Andersonand Xiang ZengBoston, USAAbstracts 161 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. We reported that neurotization, the surgical rerouting ofintact peripheral nerves originating from spinal segmentsrostral to the site of injury to distal targets, could markedlyimprove motosensory function in rats with subacutehemisection (Konya’08) or contusion SCI (Yu’13). Unlikewhat had been assumed to trigger recovery (i.e., motorcontrol rerouting) our data has engendered a novelhypothesis that central neuroplasticity including adaptingafferent signals via the newly rerouted peripheral nerve andpropriospinal projection network to invoke locomotionpattern generator may act for neural restoration. We proposeto further discuss therapeutic effect of neurotization usingdata produced by rat models of hemisection and contusionSCI, including a 2-steps strategy of neural rerouting plushuman mesenchymal stromal stem cell (hMSC) transplanta-tion after chronic contusion SCI. Our work, as a bench-siteprogress, devises a clinically feasible procedure at bothlocalized and systemic levels to treat traumatic SCI.Neurotization, as a peripheral approach of CNS repair, haslong been studied, and even sporadically applied clinically.However, lack of understanding on the mechanisms hasprevented its standardized applications. Since SCI remainsan unmet medical demand, successful development of thistreatment may greatly reduce suffering and cost for spinalcord trauma. (supported by CIMIT-DoD). Caveolin-1 alters the amyloidogenic processing ofamyloid precursor protein to amyloid-beta Rhian S. Thomas and Emma J. KiddSchool of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, U.K. Introduction: The ‘Amyloid Hypothesis’ suggests that thebuild-up of (Ab) is a primary causal event in Alzheimer’sdisease. Ab is cleaved from amyloid precursor protein(APP) by the band g-secretases, all of which have beenidentified in lipid raft-regions of the plasma membrane.Caveolae are specialised forms of lipid rafts, enrichedwith caveolin proteins. We examined whether expressionof the most ubiquitous caveolin protein, caveolin-1, couldaffect the processing of APP into Ab. Methods: Levels of caveolin-1 in astrocytoma cells weredepleted by siRNA or over-expressed by deliveringconstructs carrying the myc-tagged caveolin-1 gene tothese cells. Cells were lysed and media collected todetect intraand extracellular protein levels of lipid raftproteins, APP and APP metabolites including Ab. Results: After caveolin-1 levels were reduced by siRNAAPP levels were unaffected, however, expression levels ofAb were significantly increased Conversely, after over-expression of caveolin-1, APP levels were significantlyreduced and expression levels of Ab were unchanged. Conclusion: These results suggest that at normal physio-logical levels, caveolin-1 has a regulatory effect on Ab andmay provide a novel therapeutic target for this disease.Acknowledgements: This work was funded by BRACE. [Forposter presentation, poster board 35 on Thursday 5September 2013]. Sedimentation of cell suspensions in large diametercannulae: is it a problem?M. Trigano, E.M. Torres and S.B. DunnettParis, France and Cardiff UK Cell transplantation using embryonic cells has beenshown to be effective in animal models of Parkinson’sdisease (PD) [1,2] and Huntington’s disease, likewise inhumans for treatment of PD [3]. A major difference between animal and human therapies isthe size of the injection cannulae used. In rodent modelscells are typically delivered using small cannulae withinternal diameter of 0.159mm. In humans, larger cannulaewith internal diameters of 0.5–1.0mm will be needed andthe question has arisen as to how cell suspensions willbehave in such needles during the long surgical procedure. In this study, we investigate the behaviour of cellsuspensions using injection cannulae of 2 differentdiameters. The data show that cell sedimentation occursin both 0.5 and 0.8mm cannulae, in both vertical andhorizontal orientations severely affecting the distributionof cells in the needle in a way that would produce a veryuneven distribution of cells post injection. Acknowledgements: This work was funded by the EU FP7TRANSEURO programme. References1 Torres EM, Dowd E, Dunnett SB. Recovery of functional deficits followingearly donor age ventral mesencephalic grafts in a rat model of Parkinson’sdisease. Neuroscience 2008; 154:631–640.2 Heuer A, Vinh NN, Dunnett SB. Behavioural recovery on simple and complextasks by means of cell replacement therapy in unilateral 6-hydroxydopamine-lesioned mice. Eur J Neurosci 2013; 37:1691–1704.3 Mendez I, Dagher A, Hong M, Gaudet P, Weerasinghe S, McAlister V, et al.Simultaneous intrastriatal and intranigral fetal dopaminergic grafts in patientswith Parkinson disease: a pilot study. Report of three cases. J Neurosurg2002; 96:589–596. Comparison of mouse and human developing striatalgene expression N.N. Vinh, C.M. Kelly, A. Heuer, S.V. Precious, N.D. Allen,P.J. Kemp and A.E. RosserCardiff, UK Transplantation of neural cells derived from pluripotentstem cell sources in Huntington’s disease requires that theybe directed towards a medium spiny striatal DAPRPP-32 positive neuronal phenotype. There are published reportsof protocols to direct the differentiation of cells to an MSNphenotype that have shown success in terms of DARPP-32readout, but limited functional effect of these cells isavailable to date. This would suggest that DAPRPP-32alone is not sufficient to confirm a mature functional MSN162 NeuroReport 2014, Vol 25 No 3 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. neuron. Thus, there is a need to both understand moreabout the signals required for MSN development and toidentify a profile of markers that are indicative of normalMSN differentiation. To this end, we have carried out amolecular and histological comparison of human and mousewhole ganglionic eminence (WGE) development across theperiod during which MSNs are generated. We performedQPCR of a range of known and potential striataldifferentiation markers of WGE cells between E12 andE16 in the mouse and 6–12 weeks post conception in thehuman, and have attempted to correlate the striataldevelopment between the two species. We also performedin situ hybridization and immunohistochemical analysis ofsections over a similar developmental range for bothspecies. We carried out electrophysiological analysis ofhuman WGE cells over the same developmental period. Forthe first time, this provides key information on the geneticstages of human MSN development, and highlights somekey species differences. Vascularization for neural repair and stem celltransplantation by hyaluronan-peptide hydrogels Ying Wang, Rongkai Ju, Shanshan Li, Yujun Wen,Shukui Yu and Qunyuan XuBeijing, China Stem cell therapies are shown to be promising fordiseases or injuries to the central nervous system(CNS), but the effect is limited so far. One of thereasons is microenvironment after CNS injury is notoptimal for cell survival, especially due to the deficiencyof angiogenesis in situ. Reconstruction of a blood vesselnetwork for stem cells and injured tissue may play a keyrole for neural regeneration in the CNS. Here we have designed an angio-neurogenic hydrogel topromote angiogenesis and consequently to improve stemcell therapies for neural repair. The hydrogel was made byhyaluronan (HA), a main component of extracellularmatrix, and linked with mimic peptides of vascularendothelial growth factor (VEGF) for providing a vascular microenvironment in injured area of the CNS. Thescaffold did promote adhesion and proliferation ofendothelia and neural stem cells effectively in vitro. Also, it evoked the receptors of VEGF and promoted theangiogenesis in vivo, when transplanted in injured brain. Our results suggest that functionalized hydrogels withpeptides may provide a promising microenvironmentfor stem cells, and it should be a new approach forregenerative therapy of the injured nervous system. The development of operant delayed matching toposition for Huntington’s disease mouse models Emma Yhnell, Stephen B. Dunnett and Simon P. BrooksCardiff, UKOperant delayed matching to position (DMTP) tasks aretypically used in rats to probe spatial working memoryand usually run in standard 2-lever operant ‘‘Skinner box’’chambers. The DMTP task requires the animal to recall apreviously made left or right response following variousdelay lengths, to obtain a reward. With the development of transgene technology there isa practical need to develop these tasks for mice. As partof a larger operant test battery we aimed to achieve this,utilising the operant 9-hole box mouse chamber, withnose-poke holes replacing levers. With no delays, C57BL/6J mice learned the DMTP operanttask to >80% accuracy which decayed as a function of delay,demonstrating the viability of this task as a test of executivefunction for mice. The next phase of this work is to developthe reversal learning procedure, as both short-term memory and reversal learning are early disease signs in HD mice. Acknowledgements: This work was funded by the MRC. AAV2-mediated striatum delivery of human CDNFprevents the deterioration of midbrain dopamineneurons in parkinsonian rat and mouse model Ting Zhang, Xinmiao Ren, Xiaoli Gong, Guanzheng Hu,Wei Ding and Xiaomin WangBeijing, China Parkinson’s disease (PD) is an aging-associated neuro-degenerative disorder with progressive pathology involvingthe loss of midbrain dopaminergic neurons. Cerebraldopamine neurotrophic factor (CDNF) was recentlydiscovered to be more selective and potent on preservingdopaminergic neurons than other known trophic factors.The present study examined the neuroprotective andfunctional restorative effects of CDNF overexpression inthe striatum via recombinant adeno-associated virus type 2(AAV2.CDNF) in MPTP lesioned mice and 6-hydroxydopamine (6-OHDA) injected rats. We found that bilateralstriatal AAV2.CDNF injections, 2weeks before MPTPinjections in C57/BL6 mice, improved Rotarod behavior.AAV2.CDNF pre-treatment increased tyrosine hydroxylase(TH)-immunoreactivity in the striatum. Striatal delivery ofAAV2.CDNF 6 weeks after 6-OHDA injections in SD rats,was able to recover behavior deficits and resulted in asignificant restoration of tyrosine hydroxylase immuno-reactive (TH-ir) neurons in the substantia nigra parscompacta (SNpc) and TH-ir fiber density in the striatum.Meanwhile, long-term administration of AAV2.CDNF didn’t interfere TH expression in rat striatum. Our resultsindicate that striatal administration of AAV2.CDNF wasable to provide effective neuro-protection and neuro-restoration in the nigrostriatal system and that it may be considered for future clinical applications in PD therapy. This work was funded by National Natural ScienceFoundation of China (31000476).Abstracts 163 Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

P-153: Hyaluronic Acid Hydrogel Scaffold Containing VEGF and bFGF Affect Oestrous Cycle Restoration after Rat OvarianTissue Autotransplantation

Background: The functional longevity of ovarian transplant at present is one of the most challenging questions regarding ovary transplantation. So, we used hyaluronic acid (HA) hydrogel scaffold with combination of VEGF and bFGF for restoration of cyclical ovarian function after ovarian tissue transplantation in a rat model. Materials and Methods: A total of 25 Wistar female rats weighing betwe...

متن کامل

Neural Transplantation as a Strategy for Recovery of CNS Functions

Transplants of neural tissue have been used for nearly 100 years in order to clarify the mechanisms that underlie development and regeneration of the nervous system. With reports from Sweden and Mexico that autografts of adrenal medulla produce at least transient and perhaps dramatic improve­ment in patients severly disabled by Parkinson's disease, the techniques of neural transplantation have ...

متن کامل

The International Society for Heart and Lung Transplantation , 32 nd Annual Meeting and Scientific Sessions

Interest in pulmonary hypertension (PH) continues to grow within the International Society for Heart and Lung Transplantation (ISHLT). The PH program at the 32nd annual meeting held on 18–21 April 2012 in Prague, Czech Republic, was particularly strong, with 56 accepted abstracts and five symposia dealing with PH and right ventricular function. In this report, we will summarize selected oral ab...

متن کامل

P-80: Histological Assessment and Oestrous Cycle Restoration after Vit-rified-Warmed Rat Ovarian Tissue Autotransplantation, Using Hyaluronic AcidHydrogel Scaffold

Background: Follicular pool is reduced after transplantation of vitrified ovary. Therefore, methods should be improved to accelerate ovarian function revival and maintain follicular reserve. In this study hyaluronic acid hydrogel as a scaffold was used to improve restoration of ovarian oestrous cycle. Materials and Methods: 79 mature (~8 weeks) rats having normal estrous cycles were divided int...

متن کامل

Publication bias is present in blood and marrow transplantation: an analysis of abstracts at an international meeting.

Publication bias is the preferential publication of research with positive results, and is a threat to the validity of medical literature. Preliminary evidence suggests that research in blood and marrow transplantation (BMT) lacks publication bias. We evaluated publication bias at an international conference, the 2006 Center for International Blood and Marrow Transplant Research (CIBMTR)/Americ...

متن کامل

ذخیره در منابع من


  با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

عنوان ژورنال:

دوره 25  شماره 

صفحات  -

تاریخ انتشار 2014