Alzheimer Doctoral Scholarship
Introduction
The Hans and Ilse Breuer Foundation has awarded yearly up to three scholarships to excellent young scientists since 2006. So far, 30 scholarships holders have been funded; see details below.
The call for applications is made exclusively for doctoral projects at the DZNE and is directed to doctoral students:
- of basic research in natural science subjects such as biology, biochemistry or chemistry, but also human and veterinary medicine, and
- of health services research in the field of health sciences, human medicine, psychology, public health, social sciences, sociology, statistics and biometrics.
The scholarship consists of a basic amount of 2.300 EUR per month and a lump sum for material and travel expenses of 1.000 EUR per year.
The scholarship is paid to the DZNE to finance a doctoral position (65% of level E13). If the monthly amount of funding by the Hans and Ilse Breuer Foundation is insufficient to cover the total costs of the doctoral position, the DZNE will cover the gap.
The scholarship is granted for 36 months. The scholarship can be extended for a further 12 month upon justified application.
Application procedure
Applications must be submitted in English electronically to the Board of Trustees (info@breuerstiftung.de) by September 30th of each year. The application should consist of a maximum of five pages and be written in Arial (font size 11, line spacing 1,5). The content of the application must be based on the following model
(PDF-document, 208 KB).
Scholarship Guidelines
(PDF-document, 206 KB)
The Scientific Advisory Board (SAB) evaluates the incoming applications and makes a recommendation to the Board of Trustees. On this basis, the Board of Trustees decides on the award of the scholarships.

Publication award
Short summary of research project
Alois Alzheimer identified three key hallmarks of the disease: the presence of amyloid plaques and tau tangles, as well as increased microglia – immune cells responsible for destruction of invading pathogens in the brain. The microglial gene TREM2 was identified to play a crucial role in sustaining the microglial response in disease conditions. Using an amyloid plaque depositing mouse model of Alzheimer’s disease (AD), I found that that loss of TREM2 function increased early plaque accumulation by preventing microglial clearance of plaques. APOE, the strongest genetic risk factor for AD, plays an essential role in promoting plaque formation and its expression is mainly reported in astrocytes, another regulator of inflammation. My studies showed that not only was APOE induced in microglia around plaques, but was also strongly reduced upon TREM2 deficiency. This suggests therapeutic strategies must consider TREM2-APOE interaction as it may protectively modulate plaque clearance, but in parallel exacerbate amyloid pathology.
Parhizkar_Samira et al 2019 Nat Neurosci DZNE MUC.pdf
Professional and Higher Educational History
Name Samira Parhizkar
Date of Birth 06/16/1990
Current
Postdoc Research Associate, Washington University School of Medicine, Missouri, USA Research group: Prof. Dr. David M Holtzman
2014 – 2019
PhD, graduate studies, Ludwig Maximilians University, Munich, Germany
Summa cum laude
“Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE”
Thesis supervisor: Prof. Dr. Dr. h.c. Christian Haass2010 – 2014
MSci Neuroscience (equivalent to MSc), University of Nottingham, UK
First Class Honours
“A time-course analysis of lipopolysaccharide-induced sickness syndrome in APP/PS1dE9 mice”
Thesis supervisor: Dr. Marie-Christine Pardon2012 – 2013
Eli Lilly, Surrey, UK
“Tau-directed passive immunotherapy in a novel in vivo model of tau propagation”Thesis advisor: Dr. Michael O’Neill2011
Arthritis Research UK, University of Nottingham, UK
“Impact of joint pathology on brain function in an established model of osteoarthritic pain”
Project supervisors: Prof. Dr. Victoria Chapman, Dr. Gareth Hathway
Scholarship holders
Here you may find out which outstanding young scientists have been and are currently being supported by the Hans and Ilse Breuer Foundation. In addition to the candidates‘ CV, you may also find a project description for their respective sponsored project.
“Through my research I would like to advance the current understanding of the disease mechanisms contributing to Parkinson's disease and thus open up new therapeutic avenues.”
Project description:
The prevalence of neurodegenerative diseases increases rapidly as our population is aging and demands the urgent development of new treatments. However, in many of these diseases, it is still unknown what exactly causes the degeneration of nerve cells in the brain. In the second most common neurodegenerative disease, Parkinson’s disease (PD), researchers observe the selective death of very specialized brain cells, the dopaminergic neurons in the substantia nigra, a region in the midbrain. These nerve cells, which release the neurotransmitter dopamine, are especially important for regulating movement and thus their loss leads to typical symptoms such as tremor or slowing of movements in affected individuals.
Important clues as to what makes these cells so vulnerable, come from patients with rare, inheritable forms of PD, in which we can link a certain mutation in their genome to development of the disease. For example, a mutation in the DJ-1 gene, responsible for encoding a protein that is involved in the cellular antioxidative stress response, causes early-onset PD. In neurons of individuals with a mutation in DJ-1, we found that changes to the neurotransmitter dopamine itself was the mediator of a harmful cascade that ultimately resulted in cellular disturbances and death (Burbulla et al., Science, 2017). Interestingly, these results were also observed in neurons from patients with the much more common sporadic form of PD.
Traditionally, PD was considered a purely neuronal disease. However, recent studies suggest that alterations in other types of brain cells might contribute to neurodegeneration in PD and I am especially interested in studying how these interactions influence disease. Interestingly, DJ-1 protein is very abundant in astrocytes. Astrocytes are the supportive cells of the brain and closely communicate with neurons through a variety of signaling molecules. During my PhD project I want to study how DJ-1, and loss of its function, specifically influences the astrocyte signaling molecules and impacts the communication between astrocytes and neurons. We hypothesize that astrocytic DJ-1 could play a key role in protecting dopaminergic neurons through secreted proteins by astrocytes, and in turn, loss of DJ-1 function may contribute to PD progression, thus presenting a possible target for developing new and better medication.
To study the involvement of astrocytes in neurodegeneration, I will utilize the so-called induced pluripotent stem cell (iPSC) technology. This exciting technique allows me to convert patient’s skin cells into stem cells, which can then be turned into neurons and astrocytes in our laboratory. During my PhD, I will therefore create astrocytes from PD patient stem cells carrying a DJ-1 mutation, from sporadic PD patients and healthy subjects and compare how their signaling molecules differ. Moreover, I will investigate their effect on known PD-linked pathology in neurons and test whether treating astrocytes with drugs that affect DJ-1 levels and function can protect dopaminergic neurons from death. The results of my doctoral thesis will be an important step in further understanding the relationship of astrocyte-neuron communication and neuronal vulnerability in PD.
CV
Name Annika Wagener Birthday 29.09.1995 Place of birth Würzburg Education
Since 10/2021 PhD student | German Center for Neurodegenerative Diseases (DZNE) & Ludwig Maximilian University (LMU) Munich, Germany
“Astrocyte-neuron crosstalk in Parkinson’s disease – the impact of non cell-autonomous mechanisms in neurodegeneration”
Supervisor: Prof. Dr Lena Burbulla10/2021 – 12/2021 Visiting Researcher | Luxembourg Centre for Systems Biomedicine, University of Luxembourg
Advisors: Dr Ibrahim Boussaad and Prof. Rejko Krüger10/2018 – 04/2021 Master of Science (MSc) Molecular Biosciences with Major Neuroscience|
Ruprecht Karl University Heidelberg, Germany08/2020 – 04/2021 MSc thesis | Institute for Stroke and Dementia Research, LMU Munich
“A High-throughput workflow for CRISPR editing in human iPSCs”
Supervisor: Prof. Dr Dominik Paquet09/2019 – 02/2020 MSc Research Project | Division of Neurogeriatrics, Karolinska Institute, Sweden
“Evaluation of a novel intramembrane protease involved in Alzheimers disease”
Supervisors: Dr Simone Tambaro and Prof. Dr Per Nilsson09/2014 – 06/2018 Bachelor of Science (BSc Hons) Neuroscience | University of Glasgow, Scotland 09/2017 – 01/2018 BSc thesis | Research Institute of Neuroscience & Psychology, University of Glasgow
“Investigating memantine as a prophylactic treatment for ischaemic stroke”
Supervisor: Dr Chris McCabeHonours and Awards
10/2021 Awarded "Thiemann Visitor Programm" Scholarship by the Thiemann Stiftung Publication
Dickie, D.A., Gardner, K.*, Wagener, A.*, Wyss, A., Arba, F., Wardlaw, J. M., Dawson, J. (2019). Cortical thickness, white matter hyperintensities, and cognition after stroke. International Journal of Stroke. doi: 10.1177/1747493019851291 "I would like to contribute to integrating frailty as a promising health and age(ing) measure into German health care and research"
Project description:
Dementia is an age-associated disease. Therefore, aging and (healthy) aging in general is a topic of interest in gerontological and geriatric research. The aim is to (a) predict life expectancy and/or mortality, (b) establish a link between the aging process and (everyday) functionality, and (c) evaluate potential therapies and interventions to extend healthspan.
The predictive power of chronological age is limited and, of course, it cannot be influenced. Biological age, on the other hand, directly and indirectly affects functionality and mobility. However, the measurability of biomarkers is not easily and readily available in routine care; they require specific procedures, knowledge, and expertise.
The relationship between (biological) age, morbidity, mortality, (everyday) functionality, mobility, and health is describable and can be used both as an indicator for an intervention or as an outcome factor of an intervention in routine care. A promising concept for this purpose is frailty, a concept for describing individuals at multiple, health- and care-related levels. One of the advantages of measuring frailty compared to clinical data is the ability to derive it from existing data collected for a different primary purpose. Thus, it is possible to measure frailty retrospectively in different groups of people and in different settings without undue effort, allowing secondary analyses to investigate mechanisms of healthy aging.
In international research and care, frailty is already much more established than in Germany. Various countries have been working on a standardized implementation of frailty measurements in primary and acute care for several years. In Germany, there are different assessment instruments, but they are not used uniformly. With my PhD project I would like to contribute to the international scientific discussion and show the benefits for research with and care of people with dementia.
To achieve this, I want to develop a deficit-oriented frailty index in a guideline-based manner and apply it to existing data from a health-centered study with people with dementia (DelpHi-MV) to evaluate the usability of frailty as an endpoint and/or indicator for interventions in a second step. In the last step, a measurement tool for frailty in routine care will be tested and evaluated. For this purpose, the survey will be integrated into a study dealing with the implementation of study concepts in routine care.
After three years, a usable frailty index for the DelpHi-MV data should be available, evidence about the relationship between intervention and frailty as well as recommendations for the use of frailty in health services research studies should be elaborated.
CV
Name Melanie Boekholt Birthday 24.09.1992 Place of birth Viersen Education and professional experience
Since 2019 PhD student, German Center for Neurodegenerative Diseases (DZNE) Site Rostock / Greifswald, AG Thyrian, Interventional Health Services Research 2017 – 2019 Assistant Researcher at the Chair of Social Structure and Sociology of Aging Societies, Prof. Dr. Brandt 2016 – 2019 Master of Arts in „Aging Societies“ at the Technical University of Dortmund, Germany 2012 – 2016 Bachelor of Arts in „Gerontology“ at the University of Vechta Stipendien
2021 Summer School scholarship: “Empirical approaches to aging research” from the Institute of Gerontology, Vechta 2020 Conference scholarship for young researchers at the conference "Living and Health in Old Age" from the Institute of Gerontology, Vechta Publications (* First authorship)
Boekholt M* et al. Health care of the future - insights and strategy of (dementia) care research. JOURNAL OF GERONTOLOGY AND GERIATRICS. 2020; 53(8):735-741. Klein O*, Boekholt M* et al. Effectiveness of a digitally supported care management programme to reduce unmet needs of family caregivers of people with dementia: study protocol for a cluster randomised controlled trial (GAIN). TRIALS. 2021; 22(1):401. Kracht F*, Boekholt M* et al. Describing people with cognitive impairment and their complex treatment needs during routine care in the hospital – cross-sectional results of the intersec-CM study. BMC GERIATRICS. 2021; 21(1):425. Nikelski A*, ... , Boekholt M et al. "You have to take it that way." A study of the subjective experience of corona pandemic. JOURNAL OF GERONTOLOGY AND GERIATRIE. 2021; 54(4):359-364. Thyrian J*, Boekholt M et al. The prevalence of people with dementia in Germany - a nationwide analysis at the district level. DER NERVENARZT. 2020; 91(11):1058-1061. Thyrian J*, ... , Boekholt M et al. The situation of elderly with cognitive impairment living at home during lockdown in the Corona-pandemic in Germany. BMC GERIATRICS. 2020; 20(1):540. „I want to understand the contribution of Medin to Alzheimer's disease to expand the spectrum of possible therapeutic approaches, thereby offering new perspectives to patients and their families.“
CV
Name Marleen Veit Date of birth 15.04.1997 Place of birth Filderstadt Education
Since 11/2021 PhD Student, AG Neher, DZNE Tübingen. 10/2019 – 08/2021 Master of Science: Cellular and Molecular Neuroscience, Graduate
Training Centre of Neuroscience, International Max Planck Research School, University of Tübingen.
Project: “Pathological Effects of Medin Aggregation on the Cerebral Vasculature in a Mouse Model of Cerebral β-Amyloidosis”, AG Neher, DZNE Tübingen.10/2016 – 09/2019 Bachelor of Science: Biology, University of Tübingen.
Project: “Epigenetics of Borderline Personality Disorder and Influence of Dialectical Behaviour Therapy on the Epigenetic Regulation of FKBP5”, AG Nieratschker, University Hospital Tübingen.Other Activities
Since 05/2021 Student Initiative “Mindful Science”: Founder and Leader.
Promoting mental health in the academic context.Since 01/2021 Volunteer at Verein für Sozialpsychiatrie e.V. “Listen to the voice of those affected! I want to form a basis to consider and integrate patient preferences in dementia care”
Since September 2020, Wiebke Mohr is working as Doctoral Researcher at the German Center for Neurodegenerative Diseases (DZNE) site Rostock/ Greifswald in the RG Translational Healthcare Research. Her PhD-project focuses on patient preferences of people with dementia (PwD).
“Unfortunately, oftentimes stated preferences of PwD are not adequately considered due to the degenerative nature of the diseases. A reductionist view of PwD, which disregards their personhood, halters the implementation of person-centered care. We believe that PwD want to state preferences for their own care and should be included by participatory decision-making. Consideration of patient preferences can increase adherence to treatment and care, as well as improve interventions, which in turn would improve PwDs’ quality of life.”
Project description:
Moving towards Person-Centered Care of People with Dementia: Elicitation of Care Preferences among People with Dementia and General Practitioners
The aim of person-centered and high-value care for People with Dementia (PwD) requires that PwD must be involved in healthcare-related decision-making. Especially for PwD early participation is of utmost importance due to the progressive nature of the diseases, leading to declining levels of self-responsibility, self-determination and autonomy. Recommendations for treatment and care in national guidelines are widely based on clinical trials and expert opinions, with a focus to improve clinical parameters. Desired and undesired patient preferences and patient-reported outcomes (PROs) are rarely taken into account. Earlier preference-studies found that expert judgments often not reflect the subjective preferences of the affected patients. To enable provision of person-centered and high value care, the patient’s preferences must be considered. However, especially among PwD little is known about individual care preferences. Likewise, the preferences of general practitioners (GPs), who are gatekeepers for provision of evidence-based health care, need to be elicited. If patient and provider preferences differ, provision of value based person-centered care for PwD becomes unlikely. As of today, research that has elicited care preferences among PwD is rare and most research applied qualitative methodology. Studies, which address relevant aspects of outpatient care and support services for PwD based on stated preferences methods are scarce. Quantitative stated preference methods (i.e. Analytic Hierarchy Process) are increasingly applied in healthcare to elicit preferences for a range of healthcare services. Presently, however, data for PwD in Germany are missing. Knowledge about most and least preferred choices is essential and can increase adherence to treatments as well as improve interventions, which in turn would improve PwDs’ quality of life (QoL). When interventions are not aligned with patient’s preferences and needs, low uptake and poor adherence can waste limited resources.
My project aims to elicit PwD’s stated preferences for care, supplemented by information about the correlation with GP’s stated preferences for care of PwD. The goal is to enable provision of high-value person-centered care for PwD, and secure a cost-efficient use of scarce healthcare resources.
CV
Name Wiebke Mohr Date of Birth 12/28/1990 City of Birth Hamburg Education & International Experience
Since 09/20 DZNE, Greifswald, Doctoral Researcher Transl. Healthcare Research
Proj. Patient Preferences for Person-Centered Care among People with Dementia08/19-08/20 Syneos Health, Stockholm, Intern and SSU & Regulatory Specialist
Clinical Trials Phases II-III, Market Access Expert Certificate08/17-06/19 Karolinska Institute, Stockholm, MMSc Health Econ, Policy & Mgmt
Proj. How Managers at a Swedish University Hospital Understand Value-Based Health Care01/16-04/16 St. Francis Xavier University, Antigonish (CA), Exchange Studies
Courses in Medical Anthropology, Human Nutrition and Earth Science’s05/15-10/15 National Institute of Public Health, Copenhagen, Intern
Proj. Mapping of Health Interventions for Ethnic Minorities in Danish Municipalities09/14-06/17 University of Southern Denmark, Odense, BSc Public Health
Proj. The incorporation of Traditional Knowledge into mental health promoting practices aimed at reducing suicide among Inuit youth in Greenland10/10-07/13 University of Hamburg, Law School
Completed elementary and advanced studies, equivalent to 184 ECTSHonors & Awards
11/17-06/19 Scholarship holder, Bonn, Deutsche Begabtenförderung Publications
Nomura, S., Siesjö, V., Tomson, G., Mohr, W., et al. (2020). Contributions of information and communications technology to future health systems and Universal Health Coverage: application of Japan’s experiences. Health Res Policy. Sys 18, 73 (2020). Other activities
09/18-06/20 Academic Orchestra Royal Institute of Technology, Stockholm, Violist
Weekly 3-hours-rehearsals, concerts in major Swedish concert halls12/17-09/19 German-Nordic Political Youth Network, Stockholm, Founder & Coord.
‘Willi-Piecyk-Award’ for special European Engagement incl. a prize money of 600€08/18 Nordic WHO Simulation, Copenhagen, Delegate for Germany
Topic: Access to Essential Medicines09/06-12/15 Landes Jugend Orchester Schleswig Holstein, Kiel, Violist
Orchestra for the most talented youth musicians in Schleswig-Holstein
Link zu Volltext-Lebenslauf im Forschungsinformationssystem der Universität GreifswaldEnhancing antibody mediated amyloid-β clearance with the agonistic TREM2 antibody 4D9.
Alzheimer’s disease (AD) is a major cause of late onset dementia and is characterized by abnormal protein deposits in the brain, such as amyloid-β (Aβ) plaques and neurofibrillary tangles. Aβ has been the key focus of therapeutic research for decades, but only in recent years advances in antibody targeting strategies were showing some promising results. Specifically, the anti-Aβ antibody Aducanumab induces robust Aβ plaque clearance in the brain in clinical studies, and phase III clinical trials tentatively suggest a slowing of cognitive decline in a select group of patients. However, risk of adverse side effects due to high antibody dosing are a concern.
Other genetic factors implicated in AD point towards a prominent role for microglia, the resident immune cells of the brain. One of these genes, TREM2, codes for a microglial protein involved in potentially protective processes such as chemotaxis, phagocytosis, proliferation and energy metabolism. Microglia lacking TREM2 are unable to adjust these processes in response to Aβ, which is needed in order to adequately remove Aβ from the brain. TREM2 signaling may therefore be a target to enhance microglia-mediated Aβ clearance.
The group of Christian Haass has recently developed an antibody (4D9) that binds to TREM2 and increases TREM2 availability and signaling. 4D9 treatment was shown to reduce amyloid plaques in mice and enhance Aβ phagocytosis by cultured microglia. During this PhD project I aim to investigate whether antibody 4D9 can act synergistically with Aducanumab by dual antibody treatment in a mouse model for Aβ deposition. I hypothesize this will enhance microglia-mediated Aβ plaque clearance at lower doses of Aducanumab. In addition, I aim to assess whether dual 4D9 and Aducanumab treatment can rescue metabolic failure of microglia in the context of Aβ pathology and elucidate the molecular mechanisms of synergistic treatment on microglia through transcriptomic, lipidomic, and metabolomic profiling. As microglia are key for maintaining brain homeostasis these questions are important to consider before clinical testing in patients.CV
Name Lis de Weerd
Date of Birth 06/24/1994
City of Birth Voorburg, Niederlande
since 08/2020 PhD student, German Center for Neurodegenerative Diseases (DZNE) in Munich, AG Haass 01/2019 – 07/2020 Scientific member of staff, UK Dementia Research Institute, Cambridge, United Kingdom, AG Mallucci 09/2015 – 02/2018 Master of Science, Biomedical Sciences, Molecular Neuroscience track University of Amsterdam 01/2017 – 08/2017 Masters Praktikum Gladstone Institutes, San Francisco, USA, AG Gan Thesis: "The role of progranulin in microglial phagocytosis of apoptotic cells". 12/2015 – 07/2016 Masters Praktikum Smidt Lab, Swammerdam Institute for Life Sciences, Amsterdam, The Netherlands Thesis: "Af9 affects H3K79 methylation during cortical development". 09/2012 – 07/2015 Bachelor of Science, Psychobiology, with Honours University of Amsterdam 02/2015 – 06/2015 Bachelor Praktikum, University of Calgary, Calgary, Canada, AG Wildering Thesis: "Lateral lobe control of life-history and memory in Lymnaea stagnalis" Publications
Astrocyte Unfolded Protein Response Induces a Specific Reactivity State that Causes Non-Cell-Autonomous Neuronal Degeneration
Smith HL, Freeman OJ, Butcher AJ, Holmqvist S, Humoud I, Schätzl T, Hughes DT, Verity NC, Swinden DP, Hayes J, de Weerd L, Rowitch DH, Franklin RJM, Mallucci GR. Neuron, 2020, 105(5):855-866.e5. https://doi.org/10.1016/j.neuron.2019.12.014.
Linking the 'why' and 'how' of ageing: evidence for somatotropic control of long-term memory function in the pond snail Lymnaea stagnalis de Weerd L, Hermann PM, Wildering WC. J Exp Biol. 2017 Nov 15; 220(pt 22):4088-4094. doi: 10.1242/jeb.167395Jan Filip Tristan Hasecke
Stipendiaten 2018 "Mich treibt an, etwas zur Heilung von Alzheimer beizutragen."
Jan Filip Hasecke arbeitet seit Juni 2017 an seiner Promotion in der AG Hoyer im Institut für Physikalische Biologie an der Heinrich-Heine Universität Düsseldorf. Seine Dissertation befasst sich mit der Entwicklung eines neuen Wirkstoffes gegen Alzheimer.
„Bei Alzheimer wird angenommen, dass der Auslöser der Krankheit die Verklumpung verschiedener Proteine im Gehirn zu toxischen Aggregaten ist, welche letztlich das Absterben der Neurone auslösen. Um dieses Problem zu lösen entwickle ich in meiner Doktorarbeit eine neue Wirkstoffart: proteolytische Antikörper. Diese besitzen die Spezifität von Antikörpern, jedoch zusätzlich ein katalytisches Zentrum, wie etwa Enzyme. Diese Antikörper sollen Aggregate erkennen und gleichzeitig zerschneiden können und somit unschädlich machen. Das Besondere dabei ist, dass die proteolytischen Antikörper unverändert aus der Reaktion herausgehen und anschließend ungehindert weitere Aggregate abbauen können. Dies verspricht eine enorme Effizienzsteigerung gegenüber bisherigen Antikörpertherapien in klinischen Studien.
Besonders gefällt mir an der Arbeit, dass sie aus vielen kleinen Baustellen besteht, wodurch es stetig zu vielen kleinen Teilerfolgen kommt. Vor allem aber ist die Aussicht, tatsächlich etwas an dem Problem der Alzheimer Krankheit verändern zu können, eine treibende Kraft hinter diesem Projekt.“
Projektbeschreibung
Selektion proteolytischer Antikörperfragmente gegen Amyloid-ß via „isolation by Type Restricted Antigen Proteolysis“ (iTRAP)
Bei Proteinmissfaltungskrankheiten wie der Alzheimerschen Krankheit (AD), der Parkinson-Krankheit und Typ 2 Diabetes werden körpereigene Peptide als ursächliche Auslöser angesehen. Durch bisher nicht genau bekannte Stimuli oder Veränderungen im Gleichgewicht der Proteinhomöostase erlangen diese normalerweise harmlosen Peptide die Eigenschaft zur Aggregation. Dadurch verklumpen die Peptide, was zur Anreicherung einer Vielzahl toxischer Aggregate führt, welche resistent gegen die Abbaumechanismen des Körpers sind. In der heutigen Medikamentenentwicklung ist der am häufigsten verfolgte Ansatz zur Entwicklung einer Behandlung die Immuntherapie. Diese Therapien haben folgendes Ziel: Die Aggregate zu eliminieren, indem sie diese markieren und für körpereigene Entsorgungsmechanismen zugänglich machen. Dies soll entweder durch aktive oder passive Immunisierung erreicht werden oder durch die Verabreichung speziell entwickelter Wirkstoffe, welche an die schadhaften Peptide binden sollen. Der am häufigsten genutzte Abbau-Mechanismus ist die Markierung der Aggregate mit spezifischen Antikörpern, welche anschließend durch Mikroglia, den Fresszellen des Gehirns, aufgenommen und abgebaut werden sollen.
Leider teilen die verschiedenen Therapieansätze eins oder beide der folgenden Nachteile: I) Die Bildung von Immunkomplexen, welche zur Aktivierung des Immunsystems und einer akuten Entzündungsreaktion im Gehirn führt; II) Der simultane Abbau des therapeutischen Wirkstoffs zusammen mit den gebundenen Zielpeptiden, was eine effiziente Therapie erschwert. In meiner Doktorarbeit soll ein neuer Therapieansatz gegen Proteinmissfaltungskrankheiten vorgestellt und entwickelt werden. Dieser besteht aus hochspezifischen, proteolytisch-aktiven Antikörperderivaten, welche ihr Antigen erkennen und gleichzeitig hydrolysieren (zerscheiden) sollen. Dieser Ansatz könnte seine bisherigen Vorgänger in ihrer Effektivität fundamental übertreffen. Während die konventionellen Antikörperderivate, welche in aktuellen immuntherapeutischen Ansätzen verwendet werden, ein oder ein paar Antigene binden können, bevor sie anschließend zusammen mit ihren gebundenen Antigenen abgebaut werden, könnte ein einziges proteolytisches Antikörperderivat sein erkanntes Antigen zerschneiden, nicht nur binden, und danach noch weitere Antigene abbauen. Wodurch letztlich mehrere tausend Antigene unschädlich gemacht werden können.
Um diese proteolytischen Antikörperderivate zu entwickeln, soll die Methode: „Isolation by Type Restricted Antigen Proteolysis (iTRAP)“ etabliert werden, welche ich während meiner Zeit als Masterstudent entworfen habe. Es handelt sich dabei um eine in vitro Microbead Display Methode, welche die Anreicherung proteolytisch-aktiver Antikörperderivate aus großen Antikörper-Genbibliotheken ermöglichen soll. Dabei werden nur jene Antikörperderivate angereichert, die definierte Zielpeptide zerschneiden können. Ein besonderes Feature von iTRAP ist, dass es die Erkennung von post-translationalen Modifikationen und Konformationen innerhalb der Zielpeptide durch die proteolytisch-aktiven Antikörperderivate ermöglichen sollte. Darüberhinaus sollten die Antikörperderivate, durch den Selektionsprozess bedingt, die Fähigkeit zur Selbstfaltung und ausreichende Stabilität im reduzierenden, chemischen Milieu besitzen, um eine intrazelluläre Anwendung zu ermöglichen. Intrazelluläre Anwendungen waren bislang mit konventionellen Antikörpern undenkbar, da diese durch das intrazelluläre, reduzierende Milieu ihre dreidimensionale Struktur verlieren würden, da ihre Disulfidbrücken zerstört werden würden.
Aus diesen Gründen könnte iTRAP die Entwicklung einer neuen Generation von Therapeutika gegen Amyloid-β , Tau, IAPP, Huntingtin und andere krankheitsbezogene Peptide ermöglichen.
Lebenslauf
Name Jan Filip Tristan Hasecke Geburtsdatum 24.08.1992
Seit 2017 Promotionsstudent im Institut für Physikalische Biologie, AG Hoyer. Heinrich-Heine-Universität Düsseldorf 2014 – 2017 Master of Science in Biologie, Schwerpunkt Molecular BioMedicine, Heinrich-Heine-Universität, Düsseldorf (Gesamtnote: 1,1)
Masterarbeit: „Aggregation mechanism of amyloid-β studied by an artificial dimer“ bei Dr. Wolfgang Hoyer & Prof. Dr. Henrike Heise (Note: 1,0) 2011 – 2014 Bachelor of Science in Biologie, Schwerpunkt Molecular BioMedicine, Heinrich-Heine-Universität, Düsseldorf (Gesamtnote 1,3) Bachelorarbeit: „Characterization of the aggregation properties of a covalently linked amyloid-β dimer“ bei Dr. Wolfgang Hoyer & Prof. Dr. Henrike Heise (Note: 1,0) Auszeichnungen Dreifacher Deutschlandstipendiat Nadine Tamara Mylonas
Nadine Mylonas arbeitet seit Juli 2016 an der Graduate School for Systemic Neurosciences (GSN) und dort in der Fachgruppe von Prof. Dr. Harald Steiner, Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), München. Im Fokus ihrer Arbeit liegt die γ-Sekretase. Hierbei handelt es sich um ein Enzymkomplex, welches bei der Produktion von Beta-Amyloid eine Schlüsselrolle übernimmt. Ablagerungen dieses Proteins stellen ein Hauptmerkmal der Alzheimerschen Krankheit dar.
„Das Ziel meiner Doktorarbeit ist es nun zu entschlüsseln, wie die γ-Sekretase zu spaltende Proteine erkennt und schließlich teilt. Ein präzises Verständnis des Erkennungs- und Spaltungsmechanismus ist essentiell, um die Entwicklung und Verbesserung von Medikamenten sowie präventiven Strategien für Alzheimer voranzutreiben. Es ist überaus spannend, immer wieder neue Hypothesen aufzustellen und diese zu überprüfen, um diesen komplexen Mechanismus so besser zu verstehen. Ich hoffe sehr, dass ich mit meiner Arbeit einen Beitrag für die Erforschung der Alzheimerschen Krankheit leisten kann.
Projektbeschreibung
Alzheimer ist eine neurodegenerative Erkrankung, die 1906 erstmals von Alois Alzheimer beschrieben wurde. Die Erkrankung zeichnet sich durch Ablagerungen der beiden Proteine Beta-Amyloid (sog. Aβ-Plaques) und Tau (sog. Fibrillen) im Gehirn eines an Alzheimer erkrankten Patienten aus. Im Verlauf der Krankheit ist die normale Funktion der Nervenzellen zunehmend gestört und letztlich sterben die Zellen, sowie die Verbindungen zwischen den Nervenzellen ab. Betroffene leiden zunehmend unter Gedächtnis-, Orientierungs- und Sprachstörungen, wie auch unter einer Veränderung der Persönlichkeit.
Im Fokus meiner Doktorarbeit liegt die γ-Sekretase, ein Enzymkomplex welcher bei der Produktion von Beta-Amyloid eine Schlüsselrolle übernimmt. Bisher ist eine Vielzahl von Proteinen (Substrate) beschrieben worden die von diesem Enzym geschnitten werden können. Trotz großer Fortschritte, wie beispielsweise der Aufklärung der Struktur der γ-Sekretase, ist immer noch unklar wie die Sekretase ihre Substrate erkennt und letztendlich schneidet. Es wird vermutet, dass eine gewisse Flexibilität der Substrate wichtig ist, damit diese das aktive Zentrum der Sekretase erreichen und schließlich von dieser geschnitten werden können.
Das Ziel meiner Doktorarbeit ist es (kinetische) Parameter für die Bindung und Spaltung ausgewählter Substrate untereinander zu vergleichen um so den Erkennungs- und Spaltungsmechanismus der γ-Sekretase zu entschlüsseln. Ein zentraler Aspekt ist dabei der Einfluss struktureller Eigenschaften der verschiedenen Substrate auf diese Parameter. Die genaue Analyse des Erkennungs- und Spaltungsmechanismus von γ-Sekretase Substraten hilft uns zu verstehen wie die Substrate, genauer welche Eigenschaften, wichtig für die Erkennung und die Prozessierung durch die γ-Sekretase sind.
Zusätzlich werde ich in meiner Doktorarbeit die Bedeutung der Lipide für die Substraterkennung und -spaltung untersuchen, da nachgewiesen werden konnte, dass verschiedene Lipidklassen die Aktivität der γ-Sekretase beeinflussen können. Zudem wurde gezeigt, dass die Lipidzusammensetzung der Membran durch die Ernährung beeinflusst werden kann. Dies könnte wiederum die Spaltung der Substrate durch die γ-Sekretase beeinflussen. Unklar ist jedoch wie die Erkennung und Spaltung der Substrate durch die Lipide beeinflusst wird. Um die Rolle der Lipidumgebung für die Erkennung und Spaltung der Substrate besser zu verstehen, werde ich (kinetische) Parameter der γ-Sekretase in Modelmembranen mit unterschiedlicher Lipidkomposition bestimmen.
Ein präzises Verständnis des Erkennungs- und Spaltungsmechanismus von γ-Sekretase-Substraten ist essentiell um die Entwicklung und Verbesserung von Medikamenten, sowie präventiven Strategien für Alzheimer voranzutreiben.
Lebenslauf
Name Nadine Tamara Mylonas
Geburtsdatum 14.06.1991
seit 2016 Promotionsstudent an der „Graduate School for Systemic Neurosciences” (GSN), Arbeitsgruppe von Prof. Dr. Harald Steiner, Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), München 2015 – 2016 Praktikum bei Danone Nutricia Research, Utrecht
2013 – 2015 Master of Science in Human- und Molekularbiologie, Universität des Saarlandes, Saarbrücken 2012 – 2013 Erasmus Semester, Universität Sheffield, Sheffield 2010 – 2013 Bachelor of Science in Biologie, Ludwig-Maximilians-Universität, München Anika Reifschneider
Projektbeschreibung
Der Forschungsschwerpunkt meiner Doktorarbeit ist der molekulare Mechanismus der frontotemporalen Lobärdegeneration (FTLD). FTLD gehört wie die Alzheimer Erkrankung zu den neurodegenerativen Erkrankungen und ist zudem die zweithäufigste Form präseniler Demenz. Wie auch bei Alzheimer leiden betroffene Patienten unter der Bildung von Proteinablagerungen im Gehirn, welche, aufgrund der betroffenen Hirnbereiche, vor allem zu Sprachstörungen und auch zu Verhaltensauffälligkeiten und Persönlichkeitsänderungen führen können. FTLD kann sowohl sporadisch als auch in einer genetisch vererbten Variante auftreten. Um sowohl eine sichere Diagnostik als auch eine Therapie der Krankheit zu ermöglichen, ist es wichtig, dass wir zuerst die molekularen Mechanismen verstehen, die die Krankheit verursachen. Die Krankheits-assoziierten Gene könnten dabei ein entscheidender Schlüssel sein.
Eines dieser krankheitsspezifischen Gene ist Progranulin. FTLD relevante vererbte oder spontan erworbene Veränderungen in diesem Gen führen häufig zu einer starken Reduzierung des Progranulin Proteins (PGRN). PGRN scheint eine wichtige Rolle bei Entzündungsprozessen, der Wundheilung und dem Zellwachstum zu spielen. Es wurde außerdem beschrieben, dass ein kompletter Verlust des Proteins im Körper zu einer lysosomalen Speicherkrankheit, der neuronalen Ceroid-Lipofuszinose, führt. Zudem wurde festgestellt, dass auch in Mausmodellen der FTLD die Expression von lysosomalen Proteinen verändert ist. Deshalb wird vermutet, dass PGRN zusätzlich zu den bereits bekannten Funktionen, eine wichtige Aufgabe bei dem Abbau zellulärer Proteine hat. In meinem Projekt werde ich daher den Einfluss von PGRN auf die Aktivität lysosomaler Enzyme untersuchen. Zudem werde ich die funktionellen Konsequenzen von zwei bisher unbekannten Mutationen im GRN Gen untersuchen.
Lebenslauf
Name Anika Reifschneider Geburtstort Neuss Geburtsdatum 19.07.1991
Seit 09/2016 Promotion “Cellular and molecular mechanisms of Parkinson’s and Alzheimer’s disease and frontotemporal lateral degeneration”
Deutsches Zentrum für Neurodegenerative Erkrankungen e.V., München
AG Haass, Supervisor: Dr. Anja Capell
10/2014-07/2016 Master of Science in medizinischer Biotechnologie
Technische Universität Berlin
09/2015-05/2016 Masterarbeit: ”Characterization of elements that skew Cas9 induced double-stranded DNA break repair towards homology directed repair”
Oslo University Hospital, Department of Microbiology
01/2014-03/2014 Bachelor's Thesis: “Aptamer based detection of thrombin and streptavidin” (grade: A+)
Technische Universität Berlin, Germany
10/2011-09/2014 Bachelor of Science in Biotechnologie
Technische Universität Berlin, Germany07/2010-08/2011 Freiwilliges soziales Jahr/Entwicklungspolitischer Dienst im Ausland, Bolivien
Deutsches Rotes Kreuz in Hessen Volunta gGmbH
Projektbeschreibung
Investigating the role of aberrant protein-protein interactions in familial ALS pathogenesis using iPSC-derived motor neurons
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that selectively affects motor neurons. Approximately 5 to 10 % of cases are familial (fALS) and result from inherited genetic mutations, such as those affecting the gene Fused in Sarcoma (FUS). The FUS protein is a crucial regulator of multiple cellular functions, including stress granule (SG) formation as well as RNA binding and processing. Aggregates of FUS are a hallmark of FUS-ALS, but the role of these aggregates in disease pathogenesis is not clear. This question remains unresolved because the majority of studies have used cellular models incapable of recapitulating the complex biology of motor neurons (MNs). As a result, no therapeutics are available to prevent or slow ALS pathogenesis. Induced pluripotent stem cells (iPSCs) provide a revolutionary approach to model ALS because they can be differentiated into a theoretically limitless number of patient-specific MNs.
In order to investigate ALS in vitro, our lab previously used CRISPR/Cas9-mediated gene editing to generate isogenic WT and P525L FUS-eGFP iPSCs. In line with existing reports, P525L FUS showed preferential abnormal cytoplasmic localization, and high content imaging performed on arsenite-stressed cells linked the mutation to altered SG dynamics. Using the GFP sequence as an affinity tag, we performed a preliminary pull down experiment on iPSC-derived MNs to explore FUS interactors, and identified protein partners interacting differentially with WT and P525L FUS. Because some of the detected proteins have been associated with ALS when mutated, we consider this as a strong clue that something relevant for the disease is happening at this level, and speculate that such interactions are integral to the induction of FUS-ALS. We propose to further investigate protein-protein interactions in this model to identify which players may be crucial for disease pathogenesis.
To determine if ALS pathology is induced via gain- or loss-of-function, we will either knock down or overexpress these proteins in iPSC-derived MNs, and evaluate the impact on ALS pathological phenotypes. Our hope is to shed some light on the mechanisms involved in disease pathogenesis, thereby facilitating the discovery of novel therapeutic approaches. Since pathological aggregation of proteins is a key feature of ALS, - and FUS inclusions have been reported also in sporadic cases, indicating a common pathological denominator - it may be possible, in the future, to develop these findings into therapeutics for most ALS patients.Lebenslauf
Name Lara Marrone Date of Birth 14.03.1991
since 2016 Center for Regenerative Therapies TU Dresden (CRTD), PreDoc 2013 - 2015 Master of Science Degree in Regenerative Biology and Medicine at Center for Regenerative Therapies TU Dresden (CRTD), Dresden, Germany
2010 - 2013 Bachelor of Science Degree in Medical and Pharmaceutical Biotechnology at the Vita-Salute San Raffaele University, Milan, Italy Publikationen
Oxygen Tension Within the Neurogenic Niche Regulates Dopaminergic Neurogenesis in the Developing Midbrain
Wagenführ L, Meyer AK, Marrone L, Storch A., Stem Cells Dev. 2016 Jan 7, (doi: 10.1089/scd.2015.0214).
Brain oxygen tension controls the expansion of outer subventricular zone-like basal progenitors in the developing mouse brain
Wagenführ L, Meyer AK, Braunschweig L, Marrone L, Storch A., Development. 2015 Sep 1;142(17):2904-15, (doi: 10.1242/dev.121939)
Adoptive T-cell therapy in the treatment of viral and opportunistic fungal infections
Mancini N, Marrone L, Clementi N, Sautto G, Clementi M, Burioni R., Future Microbiology, Vol. 10, No. 4 , Pages 665-682, (doi: 10.2217/fmb.14.122)
Adriana González
Projektbeschreibung
FUNCTIONAL ANALYSIS OF PHOSPHOLIPASE D3 (PLD3) IN ALZHEIMER´S DISEASE
Alzheimer´s disease (AD) is the most common form of progressive dementia in the elderly, for which several genetic risk factors have been described. Previously, a whole-exome sequencing study identified that a rare coding variant in the phospholipase D3 (PLD3) gene confers a two-fold risk factor in the development of AD, affecting the turnover and cleavage products of the Amyloid Precursor Protein (APP). PLD3 contains two conserved HKD motifs. As a result, it has been classified as a member of the phospholipase D family, together with the well characterized phospholipases D1 and D2. However, to date, no canonical activity or substrate has been described for PLD3.
Our group has described PLD3 as a transmembrane protein transported throughout the secretory and endocytic pathway. In early endosomal compartments, PLD3 co-localizes with APP. PLD3 is then proteolytically processed to a soluble form in acidic compartments to finally reach lysosomes. Our data indicate that PLD3 transport to lysosomes is mediated via the endosomal sorting complex required for transport (ESCRT) where previous ubiquitination of lysine residues is required for sorting into intraluminal vesicles (ILVs).
In brains derived from Pld3-deficient mice no changes of APP full length levels nor its cleavage products, including amyloid beta (Aß), were found. However, microgliosis in the dentate gyrus of the hippocampus together with a depression-like behavior was observed. As part of this project our Pld3 KO mouse will be bred into an established AD mouse model (Pld3-/-5xFAD). We believe that the AD-related phenotypes of this well described mouse strain will help us to decipher the effect of the absence of Pld3 on the turnover, transport and processing of APP, by performing cell biology and biochemical approaches, e.g. culture of primary neurons and immunohistochemistry. Previous data have shown that APP is also ubiquitinated and transported to ILVs via de ESCRT pathway to further be degraded in lysosomes, therefore, ubiquitination and MVB-dependent sorting of Pld3 in cell lines and primary neurons will be also investigated.
We hypothesize that Pld3 affects the sorting of APP in late endosomal-/lysosomal compartments and thereby its proteolytic cleavage and degradation is affected leading to an accumulation of Aß plaques and further increasing the probability to develop AD.Lebenslauf
Name Adriana Carolina González Date of Birth 12.05.1988
since 2015 PhD student. Functional characterization of new membrane-associated lysosomal proteins. Institute of Biochemistry. Unit of Molecular Cell Biology and Transgenic Research. Christian Albrecht Universität zu Kiel, Germany 2012 - 2015 Master of Science Degree in Molecular Cell Biology and Neurobiology, Department of Human Biology and Human Genetics, University of Kaiserslautern, Germany
2005 - 2010 Diploma in Biology at Simon Bolivar University, Caracas, Venezuela Projektbeschreibung
Amyotrophic lateral sclerosis (ALS) is an adult-onset disorder characterized by progressive paralysis caused by the degeneration of motor neurons (MNs). Although most cases are sporadic, about 10 % are familial and caused by genetic mutations in genes such as SOD1, FUS, and C9ORF72. Transgenic mice expressing mutant SOD1 recapitulate hallmarks of ALS pathology including MN degeneration, mitochondrial dysfunction, aggregation of SOD1 protein, progressive paralysis, and shortened lifespan. Because mitochondria play a critical role in cell survival, many groups have sought to protect MNs from degeneration by protecting mitochondrial function from ALS pathology. However, paralysis in ALS patients is not the result of MNs degeneration, but, rather, degeneration of neuromuscular junctions (NMJs), which later progresses to MN loss. As a result, any therapies aiming only at preserving MN survival are likely to have no effect on ALS pathogenesis. Instead, it is essential to focus on preserving NMJ structure and function.
iPSCs are poised to revolutionize our understanding of ALS and to enable the identification of novel therapeutics. Through reprogramming, iPSCs can be derived from an ALS patient with a specific phenotype and genotype. ALS iPSC-derived MNs can then be used to recapitulate the disease pathogenesis of the donor patient. Using gene correction, isogenic iPSCs have been generated from ALS patients with mutant SOD1 and demonstrate that iPSC-derived MNs from ALS patients recapitulate ALS relevant phenotypes in vitro. Because NMJ loss is the initial cause of paralysis in ALS patients, we argue that a scalable platform for modeling NMJ dysfunction using iPSCs would be a powerful tool to identify novel ALS therapeutics.
The project aims at developing and validating a platform technology to enable NMJ-based models of ALS for compound screening. Together with the research group of PD Dr. Dr. Andreas Hermann we have designed and are manufacturing a prototype plate that connects MNs and myotubes in a highly reproducible pattern that is scalable and compatible with high – throughput screening (HTS). We propose to develop this tool and using it to generate a first – in – class model of ALS based on NMJ function. We aim at incorporating electrodes into our plates to enable HTS of NMJ function. We previously generated iPSC lines from ALS patients with mutations in FUS and C9ORF72 and are performing gene correction on these mutations to generate isogenic iPSC lines. We propose using these MNs differentiated from these isogenic iPSC lines to test, through use of our novel plates, the effects of ALS mutations on NMJ degeneration, which is directly comparable to the initial event causing paralysis in patients. Finally, we propose performing a pilot screen of 1000 known tool compounds to validate both the use of our plates and NMJ degeneration as a screening platform. The results of this project could facilitate ALS research and give insight into new therapies.Lebenslauf
Name Jessica Bellmann Geburtsdatum 17.11.1989
seit 2016 Promotionsstudent am DFG Research Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence Department iPS Cells and Neurodegenerative Disease (Dr. Jared Sterneckert) 2013 - 2015 Master of Science in "Regenerative Biology and Medicine", TU Dresden
2009 - 2012 Bachelor of Science in "Biologie", TU Braunschweig Projektbeschreibung
Mein Forschungsschwerpunkt sind die molekularen Mechanismen der Frontotemporalen Demenz (FTD) und Amyotrophen Lateralsklerose (ALS). Diese neurodegenerativen Erkrankungen sind eng verwandt mit der Alzheimerschen Krankheit. Da jedoch andere Nervenzellen vom Zelltod betroffen sind, kommt es anders als bei Alzheimer nicht zum Gedächtnisverlust, sondern zu einer fortschreitenden Veränderung der Persönlichkeit, des Sprachvermögens oder der Muskulatur. FTD und ALS sind bislang unheilbar und führen nach einigen Jahren unweigerlich zum Tode.
Auf molekularer Ebene zeichnen sich beide Erkrankungen durch eine abnorme Umverteilung und Aggregation bestimmter Proteine aus; diese Proteinaggregate können sowohl im Zytosol als auch im Zellkern von Nervenzellen vorkommen. Zwei Proteine, die in den Proteinaggregaten gefunden wurden, sind TDP-43 (TAR DNA-binding protein of 43 kDa) und FUS (Fused in sarcoma). Beide Proteine erfüllen vielfältige Aufgaben in Zellen und es ist bekannt, dass Mutationen im TDP-43- und FUS-Gen erbliche Formen der ALS und gelegentlich auch FTD verursachen können. Wie es zur pathologischen Umverteilung und Aggregation von TDP-43 und FUS kommt und wie dies zum Krankheitsbild der FTD und ALS führt, konnte bislang nicht geklärt werden.
Meine Doktorarbeit widmet sich dem Export von TDP-43 und FUS aus dem Zellkern. Im Normalzustand sind beide Proteine hauptsächlich im Zellkern von Zellen lokalisiert. Kleine Mengen an TDP-43 und FUS können aber auch in zytosolischen Fortsätzen detektiert werden, was auf den Export beider Proteine aus dem Zellkern ins Zytosol hindeutet. Über welche Transportwege TDP-43 und FUS aus dem Zellkern exportiert werden, ist noch unerforscht. Das Ziel meiner Doktorarbeit ist es, mit Hilfe von zell- und molekularbiologischen Untersuchungen herausfinden, wie der Kernexport von TDP-43 und FUS in gesunden Zellen abläuft, um dann eventuelle Export-Veränderungen im Zusammenhang mit FTD und ALS feststellen zu können. Ich hoffe, dass meine Arbeit uns neue Einblicke in die molekularen Mechanismen der FTD und ALS gewährt und einen kleinen Beitrag zum besseren Verständnis dieser Krankheitsbilder leistet.
Lebenslauf
Name Helena Ederle Geburtsdatum 19.01.1987
seit 07/2018 Sanofi-Aventis Deutschland GmbH
09/2014 – 06/2018 Promotion mit Erlangung eines GSN (Graduate School of Systemic Neurosciences) Ph.D.-Titels am BioMedizinischen Centrum der Ludwig-Maximilians-Universtität München (Arbeitsgruppe Dr. Dorothee Dormann) 2011 – 2014 Master of Science (M.Sc.) in Molecular Biosciences (Schwerpunkt Neurowissenschaften) an der Ruprecht-Karls-Universität Heidelberg 2013 – 2014 Auslandsaufenthalt für die Masterarbeit am Cold Spring Harbor Laboratory, New York, USA 2007 – 2010 Bachelor of Science (B.Sc.) in Biology (Schwerpunkt Physiologie) an der Universität Ulm Publikationen
- Ederle & Dormann: TDP-43 and FUS en route from the nucleus to the Cytoplasm.
FEBS Letter, 2017
- Khosravi et al.: Cytoplasmic poly-GA aggregates impair nuclear import of TDP-43 in C9orf72 ALS/FTLD.
Hum Mol Genetics, 2017
- Ederle et al.: Nuclear egress of TDP-43 and FUS occurs independently of Exportin-1/CRM1.
Sci Reports, 2018
- Hock et al.: Hypertonic Stress Causes Cytoplasmic Translocation of Neuronal, but Not Astrocytic, FUS due to Impaired Transportin Function. Cell Reports, 2018
- Ederle & Dormann: TDP-43 and FUS en route from the nucleus to the Cytoplasm.
Projektbeschreibung
Primary progressive aphasia (PPA) is a clinical label for a heterogeneous group of diseases and syndromes in which a neurodegenerative disorder presents with aphasic symptoms in the initial stages. To date, PPA research has focused on brain imaging, neuropsychology, and biochemical markers of the disease and virtually nothing is known about the neurophysiology of PPA. Simple language-based tasks will be administered to gain insight into the neurophysiology of language processing in PPA patients with the target of identifying different spatio-temporal signatures of language processing in the various degenerative diseases that cause PPA. The absence or degradation of signal components leads to specific disabilities (=symptoms) in language processing.
The project will use the non-invasive techniques of event-related magnetoencephalography (MEG) and electroencephalography (EEG). This neurophysiology project will be embedded as a sub-project in a large prospective study of PPA (n=200) with a multi-disciplinary team investigating neuropsychology and imaging (MRI and PET) of PPA. The specific aim of this project is to create an electrophysiological landscape of the various forms of PPA. The deeper understanding of language processing can inform mechanistic understanding of PPA subtypes and offers potential to identify biomarkers that could differentiate between the different pathologies that can cause PPA both for diagnosis and monitoring symptomatic interventions.
Furthermore, detailed information about the spatio-temporal pattern of language processing in PPA is fundamental for the future development of treatment options like speech therapy and might lead to treatment approaches with transcranial magnetic stimulation (TMS).
Lebenslauf
Name Daniel Preiß Geburtsdatum 06.10.1987
seit 04/2014 Promotionsstudent am Deutschen Zentrum für Neurodegenerative Erkrankungen in Magdeburg, AG Hirnplastizität & Neurodegeneration (Prof. Dr. Peter Nestor) 10/2011 - 03/2014 Master of Science in "Integrative Neuroscience", Otto-von-Guericke Universität Magdeburg
Masterarbeit: A Feasibility Study of Repetitive Transcranial Magnetic Stimulation to Treat Language Fluency Impairment in Primary Progressive Aphasia related to Alzheimer’s Disease bei Prof. Dr. Peter Nestor 10/2008 - 09/2011 Bachelor of Science in “Biologie“ (Schwerpunkt: Neuro- und Verhaltensbiologie), Georg-August Universität Göttingen
Bachelorarbeit: Metrische Unterschiede an Skelettelementen der unteren Extremitäten im recht-links Vergleich bei Dr. Susanne Hummel Projektbeschreibung
The role of chromatin readers in memory formation and neurodegeneration
Alzheimer’s disease (AD) is the most common age associated form of dementia. Yet, the pathology of the disease is to date not well understood on a molecular level. While some cases of AD have a genetic underpinning, the majority of the AD cases (95%) is sporadic. The most realistic approach in the short term is a treatment to delay the onset and thus improve life quality for the affected and cut treatment costs significantly. Notably, it is commonly accepted that late onset AD is a result of variable combination of genetic and environmental risk factors. This puts epigenetic mechanisms in the spotlight for the search for novel treatments, since here environmental factors are translated into molecular alterations. The precise mechanisms how epigenetic processes control memory function are however only beginning to emerge. A finding important for my PHD project is that alterations in histone 4 at lysine 12 actetylation (H4K12ac) were linked to the pathogenesis of Alzheimer’s disease (Peleg et al, 2010). How H4K12ac is regulated during memory formation is however not understood. Moreover, it is completely unclear by which signals neuronal activity is coupled to altered epigenetic gene-expression in the nucleus. In preliminary experiments we performed a proteomic screen using SILAC technology to identify proteins that specifically bind to H4K12ac in neurons. We found a number of proteins that that are known chromatin readers, hence proteins that read the epigenetic code for example by binding to specific epigenetic modifications which initiates subsequent events that control gene-expression. The most interesting proteins were the so called Bromodomain containing proteins (BRD), BRD2 and BRD4. The function of BRD proteins in the adult brain is so far completely unknown but in other cellular systems it has been shown that BRD2 and BRD4 specifically bind to H4K12ac. Further preliminary data shows that pharmacological inhibition of BRD2/4 binding to H4K12ac in the adult hippocampus enhances memory function in mice. The host lab has generated mice that allow the conditional deletion of BRD2 in mice (Brd2 cKO mice) and Brd4 cKO mice will shortly be available. In my PhD thesis I will analyse cognitive function in Brd2/4 cKO mice. To this end I will generate mice that lack Brd2/4 in excitatory or inhibitory neurons of the adult brain and subject these animals to behavior testing including the analysis of memory function (fear conditioning, water maze, T-maze, Y-maze, novel object recognition), anxiety (Elevated plus maze, open field) and sensory motor gating function (pre pulse inhibition of startle response). I will combine the behavior data with a molecular analysis of gene-expression and chromatin plasticity. During my PhD work I want to analyse Brd2/4 levels in AD, study the role of Brd2/4 in memory formation and AD. But also investigate the cellular pathways regulated by Brd2/4 during cognition, as well as the chromatin-distribution of Brd2. And further I want to analyse Brd2-dependent chromatin marks and gene-expression. In conclusion my PhD thesis is based on solid preliminary evidence and will address for the first time to role of chromatin readers in memory formation.
Peleg S, Sananbenesi F, Zovoilis A, Burkhardt S, Bahari-Java S, Agis-Balboa RC, Cota P, Wittnam J, Gogul-Doering A, Opitz L, Salinas-Riester G, Dettenhofer M, KAng H, Farinelli L, Chen W, Fischer A (2010) Altered histone acetylation is associated with age-dependent memory impairment in mice. Science 328: 753-756
Lebenslauf
Name Hendrik Urbanke Geburtsdatum 09.06.1987 in Nürnberg
seit 01/2014 Promotionsstudent am Deutschen Zentrum für Neurodegenerative Erkrankungen Göttingen, AG Epigenetik neurodegenerativer Erkrankungen (Prof. Dr. Andre Fischer) 10/2011 - 10/2013 Master of Science “Developmental, Neuro, und Behavioural Biology”, Georg-August-Universität Göttingen, mit Auszeichung
06/2012-11/2012 Hilfswissenschaftlicher Mitarbeiter, Erforschung von Lateralität und operanter Konditionierung in D. melanogaster 10/2008 - 09/2011 Bachelor of Science “Allgemeine Biologie” mit Schwerpunkt Neurobiologie und Morphologie, Georg-August-Universität Göttingen
Publikationen
Stilling RM, Rönicke R, Benito E, Urbanke H, Capece V, Burkhardt S, Bahari-Javan S, Barth J, Sananbenesi F, Schütz AL, Dyczkowski J, Martinez-Hernandez A, Kerimoglu C, Dent SY, Bonn S, Reymann KG, Fischer A (2014). K-Lysine acetyltransferase 2a regulates a hippocampal gene expression network linked to memory formation. The EMBO Journal July 2014
Projektbeschreibung
Funktionale Analyse der Phosphorylierun von LRRK2 in Morbus Parkinson mittels humaner induzierter pluripotenter Stammzellen (iPSCs)
In einer immer älter werdenden Gesellschaft steigt die Anzahl von Menschen mit neurodegenerativen Erkrankung wie Morbus Parkinson (MP) rapide an. Parkinson ist die zweithäufigste neurodegenerative Krankheit bei Erwachsenen: Einer von tausend Europäern über dem Lebensalter von sechzig erkrankt an diesem Leiden. Bei Parkinsonpatienten sterben über den Zeitraum von mehreren Jahrzehnten die Nervenzellen in der substantia nigra pars compacta, ein für den Bewegungsapparat entscheidender Teil im Gehirn, ab, die normalerweise den Neurotransmitter Dopamin produzieren dopaminerge Neuronen im Mittelhirn, mDA Neurone). Bisher erhältliche Medikamente können allerdings nur symptomatische Linderung verschaffen, indem sie den Verlust von Dopamin ausgleichen. Derzeit sind demnach keine Wirkstoffe bekannt, die die Krankheitsursache direkt angehen und verhindern können. Daher hat die Entwicklung neuer Substanzen höchste Priorität. Wir schlagen als innovative Therapiemöglichkeit die Modulierung der Aktivität der Leucine-rich repeat kinase (LRRK2) vor.
Mutationen im LRRK2-Gen sind die am häufigsten nachgewiesene genetische Ursache für MP, allerdings ist der exakte molekulare Mechanismus bisher unbekannt. Obwohl LRRK2 in den letzten Jahren im Fokus der Parkinsonforschung steht, ist nur wenig über die Funktion des Gens in Erfahrung gebracht worden. Das LRRK2-Protein wird in Nervenzellen produziert, es kann mit einer Vielzahl von anderen Proteinen interagieren und es besitzt eine enzymatische Kinase-Aktivität, katalysiert also Prozesse innerhalb einer Zelle. Bereits vor einiger Zeit hingegen ist als grundlegendes Prinzip entdeckt worden, wie Zellen ihre Proteine regulieren und an- bzw. ausschalten können, um so die verschiedenen Prozesse innerhalb einer Zelle zu steuern. Zellen erreichen dies unter anderem durch Modifikation von bestimmten Stellen im Protein, indem z. B. eine Phosphatgruppe an eine Aminosäure gekoppelt wird Phosphorylierung. Für das LRRK2-Protein wurden über dreißig dieser Stellen festgestellt, und konsistent mit dieser Idee findet man bei Patienten mit MP-verursachenden LRRK2 Mutationen abnormale Modifikationen am LRRK2-Protein, die die enzymatische Aktivität von LRRK2 verändern. Diese abnormalen Phosphorylierungen führen dazu, dass Nervenzellen im Zellkultur- und Tiermodell neuronale Phänotypen zeigen und absterben. Modifiziert man hingegen diese abnormalen Modifikationen ist es möglich, die Nervenzellen vor Zerstörung zu retten. Daher ist es wahrscheinlich, dass die beobachteten Veränderungen von LRRK2 auch beim Patienten eine wichtige Rolle in der Krankheitsentstehung Pathogenese von MP spielen.
Die exakte Funktion für die meisten der bekannten Phosphorylierungsstellen bei LRRK2 ist unklar, insbesondere bei den im Patienten betroffenen mDA Neuronen. Um besser zu verstehen, wie die Phosphorylierung von LRRK2 und die MP-Pathologie mechanistisch verbunden sind, haben wir ein in vitro Zellmodell entwickelt, welches ein frühes Stadium der MP-Pathogenese im Patienten darstellt. In diesem Modell erzeugen wir mDA Neuronen aus Stammzellen, die wir von Parkinsonpatienten hergeleitet haben (induzierte pluripotente Stammzellen, iPSCs). Die so erzeugten Zellen werden in verschiedenen Methoden dazu genutzt, um den Einfluss und die Funktion der Phosphorylierungsstellen unter verschiedenen Bedingungen wie Stress oder genetische Faktoren zu testen.
Lebenslauf
Name Michael Glatza Geburtsdatum 16.12.1985
seit 2013 Promotionsstudent am Max-Planck-Institut für molekulare Biomedizin Münster, Department Cell and Developmental Biology (Prof. Schöler) 2009 - 2013 Master of Science in "Molekulare Biomedizin", WWU Münster und Karolinska Institutet Stockholm 2006 - 2009 Bachelor of Science in "Biowissenschaften", WWU Münster und ETH Zürich mDA Neuronen, die aus patientenspezifischen iPSCs differenziert wurden, sind die Grundlage für unser in-vitro-Zellkulturmodell von MP
Bedeutung und Ziele
In den kommenden Jahren verfolgen wir mit diesem Projekt mehrere Ziele. Kurzfristig wird unser Projekt die Auswirkungen von alternativen Phosphorylierungszuständen von LRRK2 auf die Pathogenese von MP in humanen mDA Neuronen aufzeigen. Dieses Wissen wird uns helfen zu verstehen, wie die enzymatische Aktivität von LRRK2 innerhalb einer Zelle kontrolliert wird und wie diese Regulation in MP involviert ist. Zukünftig könnten diese Phosphorylierungsstellen nützliche Biomarker während klinischer Studien sein, in denen neuartige Medikamente auf ihre Wirksamkeit hin getestet werden.
Mittelfristig werden wir in der Lage sein, weitere Elemente des Netzwerks zu identifizieren, mit denen LRRK2 interagiert und reguliert wird. Die Entzifferung dieser Regulatoren und Mechanismen wird klären, wie krankheitsverursachende Mutationen die physiologische Funktion von LRRK2 stören, und inwiefern die Modulierung der enzymatischen Aktivität von LRRK2 einen therapeutischen Nutzen für den Patienten haben könnte.
Als Langzeitziel wollen wir neue Moleküle entdecken, die diese spezifischen Phosphorylierungsstellen modifizieren, um damit eine Behandlung für Patienten zu ermöglichen. Die Regulation von Kinasen wie LRRK2 ist eine bestens etablierte Therapiestrategie, die bereits sehr erfolgreich in der Behandlung von Krebs angewendet wird. Daher ist es abzusehen, dass unser Projekt zu neuen potentiellen Zielen für die Entwicklung von Medikamenten gegen MP führen kann.
Publikationen
Peer-Reviewed Articles
Derivation and expansion using only small molecules of human neural progenitors for neurodegenerative disease modeling
Peter Reinhardt, Michael Glatza, et al.
PLoS ONE 8.3 (2013) e59252. 2013
Genetic Correction of a LRRK2 Mutation in Human iPSCs Links Parkinsonian Neurodegeneration to ERK-Dependent Changes in Gene Expression
Peter Reinhardt, Benjamin Schmid, Lena F Burbulla, David C Schöndorf, Lydia Wagner, Michael Glatza, et al.
Cell Stem Cell 12.3 (Mar. 2013) pp. 354–367. 2013
Discovery of Inhibitors of Microglial Neurotoxicity Acting Through Multiple Mechanisms Using a Stem- Cell-Based Phenotypic Assay
Susanne Höing, York Rudhard, Peter Reinhardt, Michael Glatza, et al.
Cell Stem Cell (Oct. 2012) pp. 1–13. Elsevier Inc., 2012
Non-Peer-Reviewed Articles
Little Brains in a DishMichael Glatza
Edinburgh University Science Magazine 12, 2012
Drug Testing mit StammzellderivatenSusanne Höing, Michael Glatza, Jared Lynn Sterneckert, Hans Robert Schöler
Laborwelt 4, 2012
Curriculum vitae
Name Katharine Miller Place of Birth Wauwatosa, WI, USA Date of Birth April 5, 1984 2011 - present Doctor's thesis about "Molecular biological research of translational control involved in neurological disorders." at the Duncan Laboratory of the University Hospital of Hamburg-Eppendorf, Centre for Molecular Neurobiology Hamburg 2010 - 2011 Worked on the diploma / master's thesis at the University Hospital of Hamburg, Centre for Molecular Neuobiology Hamburg, Thesis: "Molecular biological research of translational control involved in neurological disorders." 2008 Bachelor of Arts, thesis: "Molecular Biology and Genetics" at the Northwestern University in Evanston, Illinois 2006 - 2010 Laboratory technician at the Dallos Laboratory at Northwestern University, School of Communication in Evanston, Illinois, USA 2004-2006 Work study on "Molecular biological research on protein interactions in the outer hair cells of mammalian cochlea" at the Dallos Laboratory at Northwestern University, School of Communication in Evanston, Illinois 2002-2004 Assistance for the collection and organisation of data about the loss of cortisone as a reaction to stress at the Mineka Laboratory at Northwestern University, Weinberg College of Arts and Sciences in Evanston, Illinois Publications
1. CEACAM16 is associated with the Tectorial Membrane and Deafness Locus DFNA4.
Jing Zheng*, Katharine K. Miller*, Tao Yang, P. Kevin Legan, Richard Goodyear, Guy Richardson, MaryAnn Cheatham, Richard J.H. Smith, Peter Dallos. Proc Natl Acad Sci, U.S A. 2011 Mar 8;108(10):4218-23. Epub 2011 Feb 22
*indicates co-first authorship2. Interaction Between the Motor Protein Prestin and the Transporter Protein Vapa.
Soma Sengupta*, Katharine K. Miller*, MaryAnn Cheatham, Peter Dallos, Jing Zheng. Biochim Biophys Acta, volume 1803 No. 7. pp 796-804. July 2010.
*indicates co-first authorship3. Interaction between CFTR and Prestin (SLC26A5).
Kazuaki Homma, Katharine K. Miller, Charles T. Anderson, Soma Sengupta, Guo-Guang Du, Salvador Aguinaga, MaryAnn Cheatham, Peter Dallos, Jing Zheng. Biochim Biophys Acta, volume 1798
No. 6. pp. 1029-40. June 2010.4. EHD4 and CDH23 Are Interacting Partners in Cochlear Hair Cells.
Soma Sengupta, Manju George, Katharine K. Miller, Khurram Naik, Jonathan Chou, MaryAnn Cheatham, Peter Dallos, Mayumi Naramura, Hamid Band, and Jing Zheng. The
Journal of Biological Chemistry, volume 284 No. 30. pp 20121-20129. 24 July 20095. Identifying Components of the Hair-Cell Interactome Involved in Cochlear Amplification.
Jing Zheng, Charles T. Anderson, Katharine K. Miller, MaryAnn Cheatham, Peter Dallos. BMC Genomics, 25 March 2009.
Project Description
TDP-43 and TranslationalControl in FTLD, ALS and Related Neurodegenerative Disorders
Frontotemporal Lobar Degeneration (FTLD) is the second most common cause of presenile onset dementia after Alzheimer’s disease. Recent groundbreaking work supports a major role for the RNA-binding protein (RBP) TDP-43 in FTLD and other major age-related neurological diseases, including Amyotrophic Lateral Sclerosis (ALS) and Alzheimer’s disease. TDP-43 is a ubiquitously expressed, largely nuclear protein in healthy cells. In disease, aberrant localization and aggregation of TDP-43 protein are found in affected neurons 1 and disease-causing mutations in TDP-43 have been identified in both FTLD and ALS patients (reviewed in 2). Nevertheless, how TDP-43 contributes to disease remains unclear.
TDP-43’s relocalization from the nucleus to the cytoplasm in disease affected neurons raises a fundamental, unanswered question: is disease due to a loss of nuclear function, gain of cytoplasmic function, or a combination of the two? Since TDP-43 exhibits nuclear/cytoplasmic shuttling ability, it is likely that TDP-43 has important cytoplasmic functions, as has been described for other nuclear RBPs, such as Sex-lethal 3. Indeed, recent genome-wide studies 4,5 revealed that TDP-43 binding sites are also found in mRNA 3’ UTRs, a key region for cytoplasmic regulation of translation by RBPs. Taken together, available data support the idea that TDP-43 may regulate translation of a subset of mRNAs and suggest that aberrant translational regulation by TDP-43 could play an important role in promoting neurodegeneration and disease.
Here I propose to investigate potential cytoplasmic functions of TDP-43 in mRNA-specific translational control in neurons. In my thesis project I aim specifically to:
- Identify mRNAs whose translation is regulated by TDP-43 in neuronal cells
- Determine which mRNAs are bona fide in vivo translational targets of TDP-43 in mice
- Analyze the translational control mechanism used by TDP-43 on its neuronal targets
To achieve these aims I will use a combination of genome-wide and molecular approaches, many of which I have already previously established during my diploma thesis. By first searching for candidates in neuronal cell culture using polysome profiling, I will be able to easily screen for neuronal mRNA targets and rule out false positives. Subsequent focused studies in mouse will allow validation of specific targets in vivo and exploration of the relevance of targets to disease. Through a combination of cell-based assays and cell-free in vitro translation methods, insight into the underlying molecular mechanism will be obtained.
Through this study, I hope to elucidate TDP-43’s effects on translation of specific neuronal mRNAs. I expect my findings to greatly enhance our understanding of TDP-43’s neuronal functions, both in health and disease. Moreover, this approach has the potential to identify new targets for therapeutic development to treat these devastating diseases.
Curriculum vitae
Name Katrin Strecker Place of Birth Heilbronn-Neckargartach, Germany Date of Birth August 20, 1985 10/2010 - present Ph.D. with Prof. Dr. Dr.h.c. Christian Haass at the Adolf-Butenandt-Institute and DZNE, Ludwig-Maximilians-University of Munich. 09/2009 - 06/2010 Master's thesis: "Investigation of the neuronal functions of the kinase S6KII in Drosophila melanogaster" with Prof. Dr. Thomas Raabe (mark: 1,0). 02/2009 - 06/2009 Internship at the University of Edinburgh/Scotland about "How do dendritic cells multitask in the face of stimulation by diverse pathogens?" 10/2008 - 07/2010 Postgraduate studies on Biomedicine at the Julius-Maximilians-University of Würzburg, Master of Science (mark: 1,0). 04/2008 - 07/2008 Bachelor's thesis: "The impact of differential surface glycosylation on the functions of dendritic cells" with Prof. Dr. Manfred Lutz, mark: 1,0. 10/2005 - 07/2008 Studied Biomedicine at the Julius-Maximilians-University of Würzburg, Bachelor of Sciences (mark: 1,6). Curriculum vitae
Name Maria Patra Place of Birth Landshut, Deutschland Date of Birth May 2, 1986 12/2010 - present Ph.D. at Adolf-Butenandt-Institute, chair for Metabolismbiochemistry, AG Parkinson´s Disease with PD Dr. Dr. Konstanze Winklhofer (IMPRS-LS PhD Programme) 04/2010-09/2010 Master's thesis at the Institute for Virology of the Technical University of Munich with Prof. Dr. Volker Bruss: "Construction and characterization of an RFP-labeled envelope protein of HBV" (mark: 1,0) 06/2008-08/2008 Bachelor's thesis at the Institute for Virology at the Technical University of Munich with PD Dr. Ingo Drexler: "Construction of recombinant MVA for differential antigen expression" (mark: 1,0) 08/2007-09/2007 Internship at the Department of Pathology at the University of Melbourne. Working topic: "β- and γ-secretase with Alzheimer's disease" 2005-2010 Studied Biochemistry at the Technical University of Munich Project Description
PARKIN as a possible link between neurodegeneration and cancer
Mutations in the Parkin gene are responsible for the majority of autosomal recessive parkinsonism. Parkin is an E3 ubiquitin ligase with a wide neuroprotective activity. It can maintain mitochondrial integrity and prevent cell death under various stress conditions. Recently, Parkin has been shown to be implicated in the mitochondrial quality control by inducing the removal of damaged mitochondria via mitophagy.
During the last few years, Parkin has also been linked to cancer, as mutations in the Parkin gene were found in several cancer types. The cancer studies suggest a role of Parkin as a tumor suppresssor gene (TSG). In contrast, studies from PD research demonstrate an anti-apoptotic effect of Parkin under stress conditions. These two observed activities of Parkin - TSG activity on the one side and neuroprotective activity on the other side - led us to ask the question, if common or different pathways are involved in these seemingly opposing activities of Parkin. One possibility might be that the two activities are regulated by different types of ubiquitination of target molecules, as Parkin is known to be an E3 ubiquitin ligase.
The expected results of this project may help to understand mechanisms related to two common disease entities of high medical relevance – neurodegeneration and cancer. Moreover, insight into the function of Parkin could be of interest to develop novel therapeutic strategies for Parkinson's disease.Curriculum vitae
Name Pia Glöckner Place of Birth Schkeuditz, Germany Date of Birth July 31, 1980 09/2007 - present Employment as research associate for Prof. T. Arendt at Paul-Flechsig-Institute for Neuroscience at the University of Leipzig 11/2006-12/2006 Internship at Zhejiang University in Hangzhou/China 01/2006-08/2006 Employment as research associate for Prof. A. Bader at Biotechnology-Biomedical Centre at the University of Leipzig 10/1999-10/2005 Studied Biology at the University of Leipzig Project work about "Molecular insights into speciation in the Agrilus viridis-complex and the genus Trachys" (Colepotera: Buprestidae)“ with Prof. M. Schlegel Diploma thesis: "Investigation of incorporation with a 3’acyl nucleotide by the Klenow-Fragment of the E.coli- DNA-Polymerase I and preparing a evolutiv method for the improvement of the Polymerase-Activity" with Prof. M. Schlegel Publications
Rohn S., Suttkus A., Glöckner P., Arendt T., Ueberham U.: Antibody-coupled polyethylenimine conjugates can specifically deliver p16INK4a DNA into neuronal cells and protect neurons from neurodegeneration. in preparation
Pavlica S., Schmitmeier S., Glöckner P., Pisconeri A., Peinemann F., Krohn K., Siegmundschulz M., Laera S., Favia P., De Bartolo L., Bader A.: Effect of native and NH(3) plasmafunctionalized polymeric membranes on the gene expression profiles of primary hepatocytes. J Tissue Eng Regen Med. 2011 Jul 13. doi: 10.1002/term.453.
Diekmann S., Glöckner P., Bader A.: The influence of different cultivation conditions on the metabolic functionality of encapsulated primary hepatocytes. Int J Artif Organs. 30(3), 192-8, 2007.
Bernhard D., Fritzsch G., Glöckner P., Wurst C.: “Molecular insights into speciation in the Agrilus viridis-complex and the genus Trachys” (Colepotera: Buprestidae) Eur. J. Entomol. 102(4), 599-605, 2005.
Curriculum vitae
Name Christina Lang Place of birth Landsberg am Lech Date of birth 28th October 1983 since 09/2010 Graduate programme “Neurodegenerative Disease Research“ (SFB 596), LMU Munich, PhD studies in the group of Prof. Dr. Christian Haass and Dr. Anja Capell, Adolf-Butenandt Institute and DZNE, Munich 10/2009 - 05/2010 Master thesis in the group of PD Birgit Luber, Institute of Molecular Pathology, Technische Universität München 02/2009 - 07/2009 Semester abroad at the Victoria University of Wellington, New Zealand; DAAD scholarship and TUM scholarship 10/2007 - 05/2010 Studies of Molecular Biotechnology, Technische Universität München; Degree: Master of Science (grade: 1.2) 08/2008 - 02/2009 Voluntary Placement at Roche Diagnostics GmbH, Penzberg 10/2004 - 09/2007 Studies of Molecular Biotechnology, Technische Universität München; Degree: Bachelor of Science (grade: 1.5) Project description
Frontotemporal dementia associated risk factors
Frontotemporal lobar degeneration (FTLD) is the second most common cause of dementia in people under the age of 65 years.
FTLD-TDP, the largest subgroup of FTLD, is characterized by ubiquitin-immunoreactive neuronal cytoplasmic or nuclear inclusions containing TAR DNA-binding protein-43 (TDP-43) as their main component. In 2006, mutations in the progranulin (GRN) gene were found to be causative for autosomal dominant FTLD-TDP. Progranulin, an ubiquitously expressed, secreted precursor protein is involved in embryonic development, wound repair, tumour growth and inflammation and discussed as a neurotrophic factor. Interestingly, all known GRN mutations are loss-of-function mutations leading to haploinsufficiency. Therefore, a possible therapeutic approach would be to increase GRN expression in FTLD patients carrying GRN mutations.
Very recently, 7p21, encoding the uncharacterized transmembrane protein TMEM106B, was detected to be a common genetic susceptibility locus for FTLD-TDP. A potential disease mechanism was suggested in which risk-associated polymorphisms at 7p21 increase TMEM106B expression which in turn increases the risk for FTLD-TDP. Furthermore, TMEM106B seems to be an important risk factor even in GRN mutation carriers implying that GRN mutations operate upstream of TMEM106B in a pathogenic cascade. However, the exact mechanism which leads to FTLD-TDP and the relationship between GRN and TMEM106B are still unknown.
In my PhD thesis, I will investigate if TMEM106B has any influence on GRN expression and therefore might it be responsible for a varying age of onset in patients with exactly the same GRN mutation. Furthermore, I will evaluate if there are any drugs which increase GRN levels in order to rescue the haploinsufficiency phenotype in FTLD-TDP patients carrying a GRN mutation.
- projects.awardForDrs.holders.holder2011b.text
Curriculum vitae
Name Sabrina Tschickardt Place of Birth Mainz, Germany Dateof Birth January 31, 1984 01/2010 - present PhD-student at the University Medical Center of the Johannes Gutenberg-University of Mainz, Institute of Pathobiochemistry in the “Molecular Neurodegeneration” group of Prof. Dr. Claus Pietrzik 01/2009 - 09/2009 Diploma thesis at Boehringer Inghelheim Pharma GmbH & Co.KG, Department CNS, Group CNS Research III, 88397 Biberach an der Riss, Germany, Topic: "Analysis of PDE9 function in neuronal systems" with Dr. Cornelia Dorner-Ciossek (Grade: 1.1*) 08/2007 - 01/2008 Exchange Student at the Århus University in Denmark 04/2004 - 09/2009 Degree programme: Biology, Johannes Gutenberg-University Mainz, Degree: Diploma (Grade: 1.1*) 04/2003 - 03/2004 Red Cross Pain Center in Mainz, Exercise in nursing service Project description
The blood-brain barrier and Alzheimer’s disease
Alzheimer’s disease (AD) is the most common neurodegenerative disorder affecting 26 million people to date and this number will increase to approximately 140 million people until 2050. AD is characterized by neurofibrillary tangles and extracellular deposits of amyloid- (Aβ) peptides. Aβ deposition in the hippocampus results in learning and memory deficits and ultimately neuronal loss, but Aβ has also been found in cerebral blood vessel walls, a symptom referred to as cerebral amyloid angiopathy (CAA). To date, AD is incurable and there have been lots of efforts for its treatment. One of it is to increase Aβ elimination from the brain, for example across the blood-brain barrier (BBB) into the blood stream.
The BBB separates the circulating blood from the central nervous system (CNS) and plays a crucial role in brain homeostasis. Furthermore, it is essential for the supply of the CNS with nutrients, but also in restricting access of many substances from the blood into the brain. The BBB is comprised of pericytes, astrocytes and endothelial cells (ECs). Tight junctions between ECs are an essential part of the BBB because they close the intercellular space between ECs and, thereby limit paracellular flux of hydrophilic molecules across the BBB. To supply the CNS with nutrients, ECs express a large number of specialized transporters and receptors. To date, few transporters and receptors have been identified in Aβ transport across the BBB, but the mechanisms are still unclear.
P-glycoprotein (P-gp), an ATP-driven efflux transporter that limits CNS penetration of drugs, was shown to mediate the transport of Aβ across the BBB in vivo and in vitro. However, some studies failed to show an effect of P-gp in Aβ transport from brain to blood. Thus, further investigation of the distinct role of P-gp in this paradigm might open new approaches for the treatment of AD.
An additional approach for the treatment of AD is the interference with Aβ1-42 production in the brain. Non-steroidal anti-inflammatory drugs (NSAIDs) have been shown to alter β-secretase activity and to selectively lower Aβ1-42 production, which is thought to play a central role in AD pathogenesis. However, NSAIDs do not cross the BBB at high enough concentrations. Recently, apolipoprotein-modified human serum albumin (HSA) nanoparticles have been shown to carry non-permeable drugs over the BBB. Using NSAIDs coupled to nanoparticles could overcome the poor BBB permeability of NSAIDs alone and, thereby modulate Aβ1-42 production in the brain of AD patients.
Curriculum vitae
Name Matthias Voss Place of birth Kiel, Germany Date of birth 09. Dezember 1984 since 10/2009 Graduate program “Protein Dynamics in Health and Disease“, Elite Network of Bavaria since 04/2009 Graduate program “Neurodegenerative Disease Research“, SFB 596, Ludwig-Maximilians University Munich since 04/2009 Graduate student, Prof. Dr. Christian Haass and PD Dr. Regina Fluhrer, Adolf-Butenandt-Institute and German Center for Neurodegenerative Diseases 2008-2009 Diploma thesis, Prof. Dr. Ottmar Janßen, Institute of Immunology, Medical Center, University of Kiel (grade: “excellent“/0.7) 2005 - 2009 Diploma program “Biochemistry and Molecular Biology“, Christian-Albrechts University of Kiel Project description
Stuctural and functional analysis of signal peptide peptidase-like (SPPL) proteasemediated intramembrane proteolysis
As the major type of dementia in the elderly, Alzheimerʻs disease (AD) is a neurodegenerative disorder associated with progressive and irreversible loss of cognitive capabilities and memory. Histologically, AD is characterized by the presence of amyloid plaques and neurofibrillary tangles within patientsʻ brain samples. Amyloid plaques are large proteinaceous aggregates of amyloid β (Aβ) peptides and generation of these Aβ peptides is implicated to trigger a cascade of molecular events leading to neuronal cell death and eventually clinically apparent AD.
Aβ peptides are generated by stepwise proteolysis of the amyloid precursor precursor protein (APP). APP is initially processed by the β-secretase BACE and Aβ peptides are released from the cell membrane by subsequent intramembrane proteolysis mediated by the γ-secretase complex. Presenilins (PS), the active protein components of the γ-secretase complex, belong to a family of intramembrane-cleaving aspartyl proteases. This family, termed the GxGD-type aspartyl proteases due to a conserved active site motif, also comprises the signal peptide peptidase (SPP) and SPP-like proteases. Given the role of the γ-secretase complex in AD pathophysiology, this protease family is of particular interest as a very promising therapeutic target.
The project will focus on the elucidation of the atomic structure of human SPPL3, a prototypic and structurally less complex member of this family, by X-ray crystallography. This will provide insight into the catalytic and mechanistic features of this protease family and given its homologous biology to PS, it will be valuable for future drug development efforts. In addition, the determinants of substrate selectivity of the family members will be analyzed by mutagenesis approaches. Strikingly, unlike PS, SPP/SPPLs are not endoproteolysed and this discrepancy will also be addressed experimentally. Hence, this work will provide critical insight into general principles of intramembrane proteolysis by GxGD-type aspartyl proteases and will contribute to our current understanding of initial events of AD pathology.References
Lettau, M., Pieper, J., Gerneth, A., Lengl-Janßen, B., Voss, M., Linkermann, A., Schmidt, H., Gelhaus, C., Leippe, M., Kabelitz, D., Janssen, O. The adapter protein Nck: Role of individual SH3 and SH2 binding modules for protein interactions in T lymphocytes. Eingereicht.
Schwarz, N., Pruessmeyer, J., Hess, M.F., Pantaler, E., Windoffer, R., Voss, M., Sarabi, A., Sechi, A., Uhlig, S., Ludwig, A.: Sequential steps of leukocyte recruitment via the transmembrane chemokine CX3CL1. In Revision.
Voss, M.*, Lettau, M.*, Janssen, O. (2009). Identification of SH3 domain interaction partners of human FasL (CD178) by phage display screening. BMC Immunol 10, 53.
Voss, M.*, Lettau, M.*, Janssen, O. (2008). Posttranslational regulation of FasL function. Cell Commun Signal 6, 11.
Curriculum vitae
Name Daniel Fleck Place of birth Stuttgart, Germany Date of birth 13. June 1981 since 2009 Graduate program “Neurodegenerative Disease Research“, SFB 596, LMU Munich. since 2099 Institute and German Center for Neurodegenerative Diseases, Munich. 2008-2009 GPhD student, Prof. Dr. Haass and Dr. Willem, Adolf-Butenandt Diploma thesis in the labs of Prof. Dr. Stevanovic, Institute for Cell Biology and Immunology, Tübingen und Prof. Dr. Haass, Adolf-Butenandt Institute, Munich. 2007 - 2009 Graduate studies, Biochemistry, Eberhard Karls University, Tübingen. 2006 - 2007 Industrial internship at F.Hoffmann-La Roche, Pharmaceuticals, Basel. 2005 - 2006 Exchange student at the Max Planck Institutes for Biochemistry and Neurobiology, Martinsried. 2002 - 2004 Undergratuate studies, Biochemistry, Eberhard Karls University Tübingen. Project description
Investigation of BACE1 expression during development and in the context of AD pathology
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disease worldwide and is characterized by progressive and irreversible dementia. Early symptoms include short term memory loss as well as subtle changes in behavior, while in later stages abilities such as language, skilled movement and recognition are affected.
The pathological features of AD are the loss of neurons and the presence of abnormal protein deposits in the brains of patients. These deposits consist either of the tau protein which forms neurofibrillary tangles within neurons or of extracellular aggregates of amyloid β (Aβ) peptides called amyloid plaques.
The latter peptides are thought to initiate a cascade of molecular events (amyloid cascade hypothesis) that eventually lead to the clinical symptoms of AD. The Aβ peptides are generated by the stepwise cleavage of the amyloid precursor protein (APP). In a first rate limiting step, the beta-site APP cleaving enzyme 1 (BACE1) cleaves APP which in a second step is then further cleaved by the γ-secretase resulting in the release of Aβ peptides into the extracellular space. Since it is the key enzyme in the generation of Aβ, BACE1 has been object of intense research and is considered a valuable drug target.
Analysis of brains of AD patients showed increased levels of BACE1 protein/activity which are thought to be responsible for increased Aβ peptide generation. Although there is some evidence that BACE1 may be modulated by Aβ and is strongly expressed especially in the vicinity of amyloid plaques, the cause for (the local) BACE1 up-regulation is unknown.
To gain new insight into the regulation of BACE1, I will address three main questions during my PhD:
- Does BACE1 protein/activity correlate with the Aβ plaque burden and disease progression? And is the up-regulation of BACE1 a consequence of the increasing plaque and Aβ burden or rather an initial event in AD pathology?
- Where in the brain and in which cellular context does up-regulation of BACE1 occur?
- With respect to the very high physiological BACE1 levels observed during development and in the first postnatal week in mice: What are the consequences in regard to production and accumulation of Aβ? And where are cells with high BACE1 levels localized?
In summary, the study of BACE1 regulation in development and AD pathology will contribute to our understanding of the initial events in AD and may be beneficial for the development of new intervention strategies.
Curriculum vitae
Name Susanne Bürger Place of birth Halle/Saale, Germany Date of birth 18.July 1981 Since 2008 PhD. student, Paul Flechsig Institute for Brain Research, University of Leipzig, Germany, thesis title: Vascular Endothelial Growth Factor (VEGF) in ßamyloidogenesis, supported by the Alzheimer Forschung Initiative (AFI). 2007 Diploma in biology (M.S.) (final grade 1.6), Friedrich-Schiller University, Jena, Germany, thesis title: Analysis of the gene expression pattern of the wiring molecules of the semaphorin-, neuropilin-, plexin- family during the murine thalamocortical development, Junior research group of neurogenetics 2000-2006 Studies of biology, Friedrich-Schiller University, Jena, Germany, Major subjects: Neuroscience, Zoology, Cell biology and Immunobiology Project description
The role of the Vascular Endothelial Growth Factor (VEGF) in processing of the β-amyloid precursor protein
Cerebrovascular abnormalities such as thickening of the microvascular basement membranes, decreased luminal diameter (Vinters et al., 1996, Claudio, 1996; Ellis et al., 1996; Kalaria and Hedera, 1995; Kalaria and Pax, 1995; Mancardi et al., 1980), cerebral amyloid angiopathy, and microvascular endothelial degeneration (de la Torre et al., 2002; Kalaria, 2000; Soffer, 2006; Thomas et al., 1996) have frequently been observed in Alzheimer patients, from which a causal relationship between vascular mechanisms and the development of sporadic Alzheimer’s disease (AD) has been hypothesized (de la Torre and Mussivand, 1993, for reviews, see de la Torre, 2008, Isingrini et al., 2009).
There are a number of studies providing evidence that the cerebrovascular degenerations in AD are related to β-amyloid (Aβ) deposition (Attems et al., 2004; Buee et al., 1994, 1997; Fischer et al., 1990; Kalaria, 1998, 2002; Mann et al., 1986; Suter et al., 2002). Aβ may cause degeneration of both the larger perforating arterial vessels as well as cerebral capillaries, which may severely affect brain perfusion and blood brain barrier (for review, see e.g., Kalaria, 2002). Moreover, Aβ peptides have been described to inhibit angiogenesis both in vitro and in vivo (Paris et al., 2004a,b), and deregulation of angiogenic factors may contribute to various neurological disorders including neurodegeneration (for review, see Ruiz de Almodovar et al., 2009). One of the key angiogenic factor, the vascular endothelial
growth factor (VEGF), a highly conserved heparin-binding protein (Sun and Guo, 2005), was originally found in vascular endothelial cells and is able to induce vascular endothelial cell proliferation, migration and vasopermeability in many types of tissue (Ferrara et al., 2003).
Increased intrathecal levels of VEGF have also been observed in brains of Alzheimer patients as compared to age-matched healthy individuals (Kalaria et al., 1998; Tarkowski et al., 2002; Yang et al., 2004) that has been correlated with the clinical severity of the disease (Ryu et al., 2009). However, the functional significance of VEGF up-regulation in the pathogenesis and progression of AD is still a matter of debate. While VEGF and other angiogenic factors were found to be enhanced in AD (Pogue and Lukiw, 2004; Thirumangalakudi et al., 2006; Vagnucci and Li, 2003; Desai et al., 2009), there is little evidence of neovascularization in AD brain but considerable cerebrovascular abnormalities and degenerations (see, e.g. Zipser et al., 2007). Only in the hippocampus of AD patients the ongoing angiogenesis resulted in increased vascular density compared with controls (Desai et al., 2009).
It is assumed that the microvascular degenerations in AD may also be the consequence of the vasoactive detrimental effects of Aβ (Schultheiss et al., 2006; for review, see e.g., Cole and Vassar, 2008). Otherwise, there are also reports that ischemia and hypofusion may trigger accumulation and cleavage of the amyloid precursor protein into Aβ, and its deposition in the brain (Bennett et al., 2000; Jendroska et al., 1995), while the mechanisms through which these pathologies affect β-amyloidogenesis are largely unknown. The upregulation of VEGF in response to hypoxic, ischemic or hypoglycemic stress (Marti et al., 1998, 2000; Stein et al., 1995; Yancopoulos et al. 2000) suggests its involvement also in processing of the amyloid precursor protein (APP). In turn, APP is also highly expressed in the endothelium of neoforming vessels (Paris et al., 2005), and inhibitors of α-and β-secretases have been reported to inhibit angiogenesis and tumour growth (Paris et al., 2005), suggesting a role of APP metabolism also during angiogenesis. Recently, VEGF has been shown to also be involved in the induction of microglial-mediated inflammation by Aβ deposits via the microglial VEGF receptor subtype Flt-1 serving as a chemotactic receptor to mobilize microglial cells (Ryu et al., 2009).
As vascular endothelial cells are also capable to express and to secrete APP (Ciallella et al., 1999), it has been hypothesized that VEGF may also be involved in formation and deposition of Aβ. In conclusion, all the observations mentioned above prompted us to address the hypothesis whether VEGF, in addition to its angiogenic, neuroprotective and neurogenic actions, may also play a role in APP processing and in formation and deposition of β-amyloid in AD.Aims
The project proposal stresses the hypothesis that VEGF up-regulated in AD, may also play a role in the progression of the disease by affecting the processing of APP.
For testing this hypothesis, the following objectives will be addressed:
Objective 1: Does VEGF affect APP metabolism in vitro?
Objective 2: Does VEGF exert its effect on APP processing in a cell-type specific manner?
Objective 3: Does VEGF exert its action on neuronal APP metabolism in an indirect manner through selectively activating endothelial cells.
Objective 4: Does VEGF induce or trigger aggregation and fibrillogenesis of β-amyloid?
Finally, at the end of the project, the possibility of a comprehensive picture about the relationship between VEGF and the increase of β-amyloid in Alzheimer's disease will help to answer the question whether the use of drugs targeting VEGF-mediated actions in the brain, represents a strategy to prevent or to treat AD.References
RESEARCH ARTICLES
1. Bürger S, Noack M, Kirazov LP, Kirazov EP, Naydenov CL, Kouznetsova E, Yafai Y, Schliebs R. (2009)
Vascular endothelial growth factor (VEGF) affects processing of amyloid precursor protein and ß-amyloidogenesis in brain slice cultures derived from transgenic Tg2576 mouse brain. Int J Dev Neurosci, 27 (6):517-23MEETING ABSTRACTS
1. S. Bürger, M. Noack, E. Kouznetsova, Y. Yafai, L. Kirazov, E. Kirazov, R. Schliebs (2009)
Vascular endothelial growth factor (VEGF) affects processing of amyloid precursor protein and β-amyloidogenesis in brain slice cultures derived from transgenic Tg2576 mouse brain.
18th European Society for Neurochemistry (ESN), 4th Conference on Advances in Molecular Mechanisms of Neurological Disorders, Leipzig, July 11-14, 2009. Abstract in: J. Neurochem. 110, Suppl. 1, 2009, pp. 40-412. S. Bürger, M. Noack, E. Kouznetsova, Y. Yafai, L. Kirazov, E. Kirazov, R. Schliebs (2009)
Vascular endothelial growth factor (VEGF) affects processing of amyloid precursor protein and β-amyloidogenesis in brain slice cultures derived from transgenic Tg2576 mouse brain.
Saxon Biotechnology Symposium, May 26, 2009, in Leipzig. Abstract in: Saxon Biotechnology, Symposium 2009, Abstracts book (Eds. A.A. Robitzki, M. Blessing, T. Züchner, M. Brand, F. Stewart, A. Beyer, E. Schäffer), Leipzig 2009 (ISBN-Nr. 978-3-00-027884-6), p. 201.3. S. Bürger, M. Noack, L. Kirazov, E. Kirazov, E. Kouznetsova, R. Schliebs (2008)
Effect of Vascular Endothelial Growth Factor (VEGF) on processing of amyloid precursor protein in brain slice and primary cell cultures derived from transgenic Tg2576 mice.
7th Leipzig Research Festival for Life Sciences, 12. Dezember 2008. Abstract in: 7th Leipzig, Research Festival for Life Sciences Abstract Book (J. Thiery, A. Beck-Sickinger, T. Arendt, Hrsg.), Leipzig (ISBN-Nr. 3-9810760-4-4), S. 11.Curriculum vitae
Name Veit Samuel Althoff since 2009 PhD student in the workgroup of Prof. Dr. G. Multhaup
2008-2009 Employment as research associate for the SPR/MALDIcorefacility at the Institute of Chemistry and Biochemistryof the FU-Berlin in the workgroup of Prof. Dr. G.Multhaup. 2006-2008 Master of Science in Biomedicine at the University ofWürzburg. Thesis: „ Examination about the processing of the Dihydrolipoamid Dehydrogenase (DLD) signal peptide”
AG Prof. Dr. M. Zimmer, Institute of Biochemistry andPathobiochemistry, Würzburg2002-2005 Bachelor of Science in Biomedicine at the University ofWürzburg. Thesis: „Genotyping of SNPs in the human FZD3-gene ina cohort of shizophrenic patients“
AG Prof. Dr. M. Zimmer, Institute of Biochemistry andPathobiochemistry, Würzburg.Project description
Generation of Aβ peptides and characterization of their molecular
interactions in the formation of toxic oligomersSUMMARY
The amyloid-β precursor protein (APP) is first cleaved by the β-site APP cleaving enzyme (BACE) and then subsequentially processed by the γ- secretase complex to generate amyloid-β (Aβ) peptides of varying length. Aβ peptides represent the main culprits in the pathogenesis of Alzheimer disease (AD). Earliest aggregates easily self assemble into fibrils and are deposited in plaques.
There is increasing evidence that soluble low-n Aβ42 oligomers play a crucial role early in the pathogenesis of AD, as they were described to be neurotoxic and to cause cognitive deficits long before amyloid plaques are detectable. However, the exact toxic mechanism remains unclear.
In this project two different aspects will be analyzed. The sequential cleavage mechanism of the γ-secretase which leads to the generation of Aβ42 peptides will be elucidated on the molecular level. The aims are to investigate how toxic Aβ oligomers are generated during and/or after the processing of APP by the γ-secretase and how the peptides of different length are released from the γ-secretase complex.
In addition, the hydrophobic interactions of the resulting Aβ-peptides which leads to the aggregation of Aß peptides into oligomers and the association of these oligomers with lipids and/or potential membrane-bound receptors will be analyzed. The biochemical analysis of Aβ-Aβ or Aβ-lipid interactions will provide new insight into the mechanisms how these peptides mediate the toxicity on the molecular level.
The identification of the molecular interactions of toxic oligomers will open up a new avenue to identify and to combat disease causes in AD.
Curriculum vitae
Name Nambirajan Govindarajan Date of birth 24/05/1982 Place of birth Kolkata (India) since 2007 Doctoral work (PhD) at Laboratory of Aging and Cognitive Diseases, European Neuroscience Institute, Goettingen, Germany. Supervisor: Dr. Andre Fischer
2003-2005 M.Sc. Molecular Biology, International Max Planck Research School, Goettingen, Germany. 2000-2003 B. Sc. (Hons.) Human Biology, All India Institute of Medical Sciences (AIIMS); New Delhi, India Project description
TO ANALYZE THE MOLECULAR AND CELLULAR MECHANISMS BY WHICH HDACs REGULATE NEURONAL FUNCTION DURING COGNITION AND ALZHEIMER’S DISEASE
SUMMARY
The term epigenetics is most commonly defined as heritable changes in gene expression that cannot be explained by the DNA sequence. The two most studied epigenetic phenomena are DNA-methylation and Histone-tail modifications.
The DNA is wrapped around a complex of eight Histones (dimers of H2A, H2B, H3, H4) to form the basic unit/level of chromatin structure. The basic amino-terminal tails of histones carry diverse post- translational modifications, for example acetylation, methylation and ubiquitination, which build up discrete pattern of chemical marks recognized and bound by other proteins. This idea is often referred to as “histone code”. Lysine acetylation of H3 and most H4 sites leads to the relaxation of chromatin thereby making the DNA accessible for DNA binding proteins such as transcription factors. Histone acetylation is therefore generally considered to promote gene expression. The acetylation of histones is regulated by histone-acetyl transferases (HAT) and Histone-deacetylases (HDAC) that transfer or remove acetyl-groups on specific lysine residues on Histone-tails, respectively.
Recent studies from our group and others indicate that epigenetic mechanisms, namely the inhibition of HDACs, could reinstate learning behavior but most importantly also re-establish access to long-term memories in animal models for neurodegeneration (Fischer, 2007) (Alacorn, 2004) (Beglopoulos, 2006). Notably, inhibition of HDAC activity has also been implicated with improving cognitive function in wild type mice (Levenson, 2004).
However, to date little is known about the role of HDACs in neuronal function and learning and memory. In this proposal I plan to unravel the underlying molecular mechanisms by which epigenetic processes such as HDAC activity affect synaptic plasticity, learning and long-term memory. I am particularly interested in elucidating which of the 11 human zinc-dependent HDACs might be most important for the observed beneficial effects on neuroplasticity. I feel that my research will significantly contribute to further our understanding about the mechanisms underlying learning and memory and may eventually also help to develop therapeutic strategies to treat cognitive disorders such as Alzheimer’s disease (AD).
References
- Alarcon, J. M., Malleret, G., Touzani, K., Vronskaya, S., Ishii, S., Kandel, E. R., and Barco, A. (2004). Chromatin acetylation, memory, and LTP are impaired in CBP+/- mice: a model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration. Neuron 42, 947-959.
- Beglopoulos, V., and Shen, J. (2006). Regulation of CRE-dependent transcription by presenilins: prospects for therapy of Alzheimer’s disease. Trends in Pharmacological Sciences 27, 33-41.
- Fischer, A., Sananbenesi, F., Pang, P. T., Lu, B., and Tsai, L. H. (2005). Opposing roles of transient and prolonged expression of p25 in synaptic plasticity and hippocampus-dependent memory. Neuron 48, 825-838.
- Levenson, J. M., O'Riordan, K. J., Brown, K. D., Trinh, M. A., Molfese, D. L., and Sweatt, J. D. (2004). Regulation of histone acetylation during memory formation in the hippocampus. J Biol Chem 279, 40545-40559.
Curriculum vitae
Name Matthias Fassler Place of birth Jena Date of birth 28/10/1980 since 2005 PhD student at the Leibniz Institute for Age Research - Fritz Lipmann Institute, JenaResearch group: “Membrane Trafficking of Proteins involved in Alzheimer’s Disease”PhD thesis: “Understanding the role of Rer1 in the assembly of the g-secretase complex”.
2002/2003 Teaching assistant, FSU Jena 2002 Student research assistant, FSU JenaPeptide synthesis of artificial ligands of SH2 domains 2002-2005 Study of Biochemistry and Molecular Biology at the Friedrich Schiller University Jena, GermanyElective intensive course: Molecular medicineGraduation as Diplom-BiochemikerGrade: 1.3 (A-)Diploma thesis: “Studies on the molecular interaction of recombinant scorpion a-toxins with voltage sensors of sodium channels”
1994-1999 Grammar school “Carl Zeiss” for mathematically, technically, scientifically gifted childrenAbitur grade (final secondary school examinations): 1.0 (A) Thesis abstract
Alzheimer’s disease (AD) is the most frequent form of dementia found in the elderly. The Aβ peptides that are the principal component of the amyloid plaques in the brains of AD patients are released to the extracellular space by the activity of g-secretase. The g-secretase is a multimeric, high-molecular-weight complex composed of presenilin (PS), nicastrin, anterior pharynx-defective phenotype 1 (Aph1) and PS enhancer 2 (Pen2). In previous work a quality control mechanism for g-secretase assembly was suggested that is based on retention signals, which keep unassembled subunits and assembly intermediates in the endoplasmic reticulum (ER). These signals become inactivated in the properly assembled complex and the g-secretase is transported to the plasma membrane and the endosomes where it is active. It was the aim of the present study to provide evidence for the proposed quality control hypothesis for g-secretase assembly. Read more
Project description
Summary PhD thesis: “Understanding the role of Rer1 in the assembly of the g-secretase complex”
g-secretase is a high molecular weight complex composed of Presenilin (PS), Nicastrin, Aph1 and Pen2. The assembled g-secretase shows pro- tease activity and cleaves several substrates like the amyloid precursor protein (APP) within their transmembrane domain. Thus, g- secretase mediates the final step in the production of the amyloid b-peptide, the principal component of the amyloid plaques in the brains of Alzhei- mer´s disease patients.
The g-secretase complex assembles in the endoplasmic reticulum (ER) and is exported to its sites of activity. A quality control system ensures that only fully assembled g-secretase leaves the ER. We hypothesize, that this quality control mechanism is based on ER-retention/retrieval signals on every g-secretase subunit, which keep the unassembled sub- units in the ER. Upon assembly these signals are supposed to be mas- ked and the subunits are no longer substrate of proteins that facilitate the retention/retrieval.
We previously identified ER- retention/retrieval signals in PS1 and Pen2, both located in transmembrane domains. We also identified a protein, Rer1, which mediates the retention/retrieval of Pen2 via transmem- brane interactions. Thus, we suppose that Rer1 might in- fluence the assembly of g-secretase via the Pen2 subunit.
In the proposed project we want to elucidate the molecular mecha- nisms of g-secretase complex assembly and analyze the role of Rer1 in this process. A siRNA screen to find interaction partners for transmembrane-based retention will be performed. To this end, we will utilize reporter constructs, which undergo Rer1 mediated ER- retention/retrieval as a sensor for the integrity of the Rer1-based quality control mechanism. Cells stably expressing the reporter constructs will be reversely transfected with a siRNA library. Subsequently, the localization of the reporter con- structs will be analyzed using high content microscopy. Additionally, we will screen for direct interaction partners of Rer1 using a yeast 2-hybrid screen based on a human brain cDNA library. It is likely, that we will find proteins in the screens that affect the described quality control mechanism for g-secretase assembly. These proteins will be analyzed further to determine their exact role in the complex assembly of g-secretase.
Curriculum vitae
Name Martin Siepmann Place of birth Münster, Germany Date of birth 08/05/1977 since 2005 Ph.D. Student, Molecular Cellbiology AG prof. Dr. Jochen Walter, University of Bonn 2003/2004 Diploma thesis "Functional Analysis of the middle domain of the molecular Chaperone Hsp90 with molecularbiological and biochemical methods", Protein Folding Group AG Dr. Wolfgang Obermann, Institute for Genetics, university of Bonn 1998-2004 Universities of Darmstadt and Bonn, Diploma in Biology Project description
Molecular Mechanisms in the Regulation of Neprilysin
Accumulation of the amyloid ß-peptide (Aß) is a common neuropathological feature of Alzheimer`s disease (AD). Aß derives from proteolytic processing of the ß-amyloid precursor protein (ßAPP) involving at least two distinct membrane bound proteases. Aß levels in the brain are not only determined by the activity of proteases involved in its generation, but also by enzymes able to degrade this peptide. Several zinc-metalloproteases have been shown to cleave Aß, including the insulin degrading enzyme, the endothelin-converting enzymes-1 and -2 and the type II membrane protein neprilysin (NEP).
Accordingly increased expression of NEP activity results in enhanced degradation of Aß and decreased Aß deposition.
This PhD project aims to identify molecular mechanisms that regulate the subcellular trafficking and activity of NEP. By using biochemical and cell biological approaches, specifically the role of phosphory- lation in the regulation of NEP metabolism and NEP localisation are investigated. Initial studies demonstrated that NEP is phosphorylated in vitro and in vivo and I am now focusing on the characterisation of the underlying signalling pathways, including the identification of protein kinases that phosphorylate NEP and the respective phosphorylation sites.
Since previous investigations showed that somatostatin signalling affects NEP activity and localisation, the involvement of NEP phosphorylation in these processes will be investigated. This research project should lead to novel insights into the regulation of NEP, its Aß degrading ability and allow the identification and evaluation of potential approaches to decrease Aß levels in the brain as therapeutic strategies for the treatment of Alzheimer's disease.Curriculum vitae
Name Lena Bouman Place of birth Heidelberg Date of birth 18.01.1979 since 2005 PhD under K. F. Winklhofer, MD PhD at the Adolf-Butenandt Institute, Ludwigs-Maximilians-University, Munich.
2003/2004 Diploma thesis in the laboratory of Prof.J. Darnell at the Rockefeller University, New York, USA (Supervisor: M. Henrikson, PhD) 2000-2001 Erasmus-exchange at the Université de Paris XI (France) 1998-2001 Undergraduate studies at the Phillipps-University Marburg
Project description
Role of Parkin in Stress Response Pathways
Parkinson’s Disease (PD) is a movement disorder characterized by a progressive loss of dopaminergic neurons in the substantia nigra pars compacta. The recent identification of gene mutations responsible for familial PD may advance our understanding of the molecular mecha- nisms underlying neuronal degeneration. Among the genes which are responsible for monogenic familial variants of PD, the parkin gene (PARK2) seems to play a prominent role.
The parkin gene encodes a 465 amino acid protein with an ubiquitin- like domain (UBL) at the N-terminus and two RING finger motifs close two the C-terminus, suggesting that parkin has an E3 ubiquitin ligase activity. Various putative parkin substrates have been reported, however these proteins are highly diverse and so far did not contribute to understand the physiological role of parkin.
We and others observed that parkin has the capacity to protect cells against different stress conditions, including oxidative stress, mito- chondrial and endoplasmic reticulum (ER) stress, however the underlying mechanism is still elusive. My PhD project is focussed on the mechanism of this neuroprotective activity. The goal of the study is to understand the regulation of parkin activity and to identify signaling path- ways involved in the cytoprotective action of parkin. Specifically, I want to address the question of whether parkin can mediate an inter- organelle crosstalk between mitochondria and ER.
Parkin can protect cells from both mitochondrial and ER stress and both organelles play an important role in mediating pro- and anti-apoptotic pathways in response to cellular stress. Based on the protective effect of parkin against ER stress, a possible effect of parkin on ER stress pathways (unfolded protein response, ER-associated degradation) will be addressed.
Insights into the physiological function of parkin and the identification of signaling pathways modulated by parkin may serve as a basis for the development of new therapeutic strategies, which could halt or delay the progression of PD.
Curriculum vitae
Name Sascha Norbert Lange
Place of birth Hamburg Date of birth 20.11.1970 since 2006 PhD student at the Institute for Neuropathology, University Medical Center Hamburg-Eppendorf
2005/2006 Research associate, Bone marrow transplantation unit, University Medical Center Hamburg-Eppendorf 2004/2005 B. Diploma thesis at the Centre for Molecular Neurobiology Hamburg
Institute for Molecular Neuropathology2000-2004 Studies in biochemistry and molecular biology in Hamburg
1997-2000 Vocational training as biology lab assistant at the Fraunhofer Arbeitsgruppe für Toxikologie und Umweltmedizin, Hamburg
Project description
Mechanisms of protein degradation in dementia
Currently in Germany over one million people suffer from dementia. According to estimates by the World Health Organisation, dementias such as Alzheimer´s Disease the fourth most common cause of death among people older than sixty years.
The exact causes for dementia are poorly understood.However, it is assumed that dementias share common pathways. Neuronal accu- mulation of malprocessed proteins is the hallmark of a number of neurodegenerative disorders such as diffuse Lewy body disease, tauopathies and familial encephalopathy with neuroserpin inclusion bodies (FENIB). Accumulation of malprocessed proteins is a dynamic process, resulting in the disturbed balance between synthesis and degradation. The result is protein aggregation in the cells, as the cause of a subsequent dementia. The familial encephalopathy with neuro- serpin inclusion bodies (FENIB) is an inherited disease of the brain that causes demantia already in the early stages of life.
A protein that is very similar to the neuroserpin is found in the nematode Caenorhabditis elegans (C. elegans). Through changes in the naturally occurring protein, it is possible to mimic the disease and analyse it. In the living worm single proteins can be switched off and thus signal paths, that might play a role in neuroserpin-interaction be elucidated. In the living organism it is also possible to test certain substances that prevent or delay the beginning of the aggregation.
The objective of my PhD thesis is to identify components involved in the degradation of aggregated neuronal proteins. For this, we have gene- rated a C. elegans based model of FENIB employing fluorescently tagged, aggregation-prone forms of neuroserpin. This allows for high throughput screens and subsequent isolation of new genes and pharmacological substances modulating degradation of aggregated neuroserpin. These studies will be extended to mammalian cell culture and in a murine model system. Data from these studies will be valuable in identifying pathways of protein degradation in neurodegeneration.
Curriculum vitae
Name Roja Barikbin Place of birth Elmshorn, Germany Date of birth 29.08.1979 since 2007 PhD in the field of neuronal protein degradation with the subject "C. elegans as a model system to analyse Ataxin-3/ATX-3-dependent neurodegeneration". The work is carried out in the lab of Dr. Thorsten Hoppe at the Center for Molecular Neurobiology Hamburg (ZMNH), University of Hamburg since 2007 Postgraduate studies in molecular biology at the University of Hamburg 2005/2006 Diplomatheses in the field of neurobiology with the subject "Identification of interaction partners and functions of the protein ATX-3/Ataxin-3 within the ubiquitin proteasome system in C.elegans". The work was carried out in the lab of Dr. Thorsten Hoppe at the Center for Molecular Neurobiology Hamburg (ZMNH), University of Hamburg. Grade: "sehr gut" (highest possible grade) 2000-2005 Study of biology at the University of Hamburg. Main subject: Human biology. Minor subjects: applied botany and cultural anthropology Project description
Molecular Mechanisms of Spinocerebellar Ataxia Type 3 (SCA3)
The neurodegenerative disease spinocerebellar ataxia type 3 (SCA3) is the most common dominantly inherited ataxia. SCA3 belongs to a group of at least nine polyQ (polyglutamine) ailments.
These polyQ disorders are based on the pathological accumulation of the amino acid glutamine within the polypeptide, which causes mis- folding and subsequent accumulation of the insoluble disease protein in neurons. The pathogenesis of SCA3 is slowly progressive with a late onset of dysfunctions impairing postural balance and coor- dination of all body movements, including gait, speech and fine movements of the hands.
Mutations in the ataxin-3 gene are in most cases responsible for inherited forms of SCA3. The Ataxin-3 protein is a ubiquitin-specific protease; however, its precise role throughout the ubiquitin- proteasome system (UPS) is not clear at present. In this context it is noteworthy that several different neuro- degenerative diseases, such as SCA3, are associated with defects within the UPS.
The aim of my PhD thesis is to gain insights into the molecular mechanisms that underly the SCA3 disease using Caenorhabditis elegans as a model organism. I will combine biochemical and genetic approaches to identify new factors involved in the pathogenesis of this polyQ disease. Furthermore, functional in vitro and in vivo studies may help to elucidate the physiological role of the Ataxin-3 protein. A prime objective is to establish a disease model for SCA3 in C.elegans by per- forming complementation studies with human Ataxin-3 and the C.ele- gans Homolog ATX-3 to understand the interplay between Ataxin-3 mutations and protein aggregation.
Curriculum vitae
Name Sonja Zilow Place of birth Heidelberg, Germany Date of birth 30.12.1980 since 2005 PhD under Prof. Haass at the Adolf- Butenandt- Institute,
Ludwig-Maximilians University, Munich2005 Masterarbeit: “Analysis of the role of Rer1 in ER-retention of non-assembled gamma-secretase complex components” am Adolf- Butenandt- Institut, Ludwig-Maximilians Universität, München (AG: Prof. Haass, Dr. Kaether) 2004/2005 Biotechnology, Master of Science at the University of applied science, Mannheim 2000-2004 Biotechnology, Bachelor of Science with Honours at the University of Applied Science, Mannheim Project description
Identification of proteins binding to the 5`untranslated region of the b-secretase
Alzheimer`s disease (AD) is the most common form of dementia among older people. It is a progressive, neurodegenerative disease characterized by neuronal loss, astrogliosis, amyloid plaques and neurofibrillary tangles in the brain.
Plaques mainly consist of the 40-42 amino acid long amyloid-b peptide (Ab) which is derived through proteolysis of the b-amyloid precursor protein (APP) by b- and g-secretase. In contrast a-secretase prevents the formation of Ab by cleaving APP within the Ab domain.
Analysis of brains of AD patients showed that BACE1 protein expression in comparison to brains of patients with no AD is increased by a factor of 2.7. This increase in BACE1 protein expression is not based on higher mRNA levels. Therefore it seems that BACE1 is regulated through post-transcriptionally mechanisms such as translational control or reduced degradation. Recently, it was shown that the 5`untranslated region (5`UTR) of BACE1 inhibits the translation of the BACE1 mRNA but not the transcription of BACE1. A possible explanation for the ob- served effects of the 5`UTR of BACE1 on the protein expression is that proteins which bind to the 5`UTR may be responsible for the repression of BACE1 translation.
Since BACE1 expression plays a crucial role in the pathogenesis of AD, the aim of the current project is to understand how the expression of BACE1 is regulated in more detail. Therefore the goal of my PhD thesis is to identify proteins binding to the 5`UTR of BACE1 since we hypo- thesize that these binding proteins might regulate translation of BACE1.
The identification and characterization of these proteins should allow a better understanding of the regulation of BACE1 expression in AD as well as in healthy people. This can lead to the identification of signal transduction pathways that might play a role for the increased BACE1 expression in AD. The better understanding of the BACE1 expression might open completely new strategies for drug development.
Curriculum vitae
Name Simone Back since 2005 Ruprecht-Karls-University of Heidelberg, Germany, PhD thesis in the research group of Prof. Dr. Dr. h.c. K. Beyreuther at the Center of Molecular Biology Heidelberg (ZMBH), Germany
Title: Analysis of the influence of PAT1a and RME-6 on APP endocytosis and processing.2004/2005 Teaching Ruprecht-Karls-University of Heidelberg, Germany, Degree dissertation in the research group of Prof. Dr. Dr. h.c. K. Beyreuther at the Center of Molecular Biology Heidelberg (ZMBH)
Title: Analysis of APP subcellular transport in neurons1999-2005 Ruprecht-Karls-University of Heidelberg, Germany, Degree student in biology. Main subjects: molecular biology, cell biology and zoology, Final grade: 1,3 (=First class) 1990-1999 High school leaving certificate, Carl-Benz-Gymnasium in Ladenburg
Final grade: 1,8 (=First class)
Project description
Project proposal
One of the main characteristics in Alzheimer´s disease are senile plaques. They are mainly composed of a short peptide (b-Amyloid, Ab) derived from the amyloid precursor protein (APP). APP is cleaved by either the a-secretase or, alternatively, by the b-secretase followed by the subsequent cleavage of g-secretase, which leads to the release of Ab or the non-amyloidogenic p3, respectively. As the different secretases are located in distinct subcellular compartments, a change in APP trafficking affects the time that APP spends together with α- or β-secretase in a common compartment and thereby influences its processing. Therefore it is of great interest to investigate the molecular mechanisms underlying the control of APP transport.
For the regulation of cellular transport processes intracellular protein sorting motifs play a crucial role. For APP two of such motifs are known: the 665YTSI668 and 692NPTY695. In a recent study we showed that PAT1a (Protein Interacting with APP Tail 1a) interacts with one of the two putative endocytosis motifs, 665YTSI668(BaSS), of APP and thereby affects APP surface levels as well as APP processing. Recently, we identified a Rab5 GDP/GTP exchange factor, RME-6, as a novel putative binding partner of PAT1a, possibly linking APP to the Rab5 dependent transport machinery of the cell.
Within my PhD thesis I investigate the influence of PAT1a and RME-6 on APP transport and sorting in the cell. Therefore I work on the one hand with cell types showing no asymmetric organization (non-polar cells) and on the other hand with polar cells e.g. nerve cells containing on both axons and dendrites as cellular protrusions. These studies will give deeper insights in the molecular mechanisms of transport, sorting and processing of APP and will thereby contribute to a better under- standing in cellular mechanisms forming the basis of the formation and progression in Alzheimer´s disease.Curriculum vitae
Name Ali Taghavi Place of birth Date of birth 03.09.1980 since 2006 PhD in Medicinal Chemistry; TUD, Darmstadt. AK: Prof.
2004/2005 B.Sc in Medicinal Chemistry; KTH, Stockholm. 2003/2004 M.Sc in Organic Chemistry; Ecole Polytechnique, Paris. 1998-2003 B.Sc Chemical Engineering; KTH, Stockholm.
Project description
Project 1:
Although amyloid proteins feature each a unique amino acid sequence, they all share very similar ultrastructural and physicochemical properties. Inclusions of amyloid proteins in the brain are associated with diseases such as Alzheimer’s disease, Parkinson’s disease and Creutzfeldt-Jakob disease. The hallmarks of AD are Senile Plaques (SPs) and Neurofibrillar Tangles (NFTs). However, both SPs and NFTs occur in over one hundred different related diseases. Therefore, a tremendous value would be added, both for diagnosis and treatment, to a ligand that selectively binds to one form of these amyloidic lesions only.
Efforts to develop imidazo[1,2-a]pyridines (IMPYs) as radiotracers for amyloid imaging have generated several lead compounds. E.g. [123I]IMPY, [18F]FPM-IMPY, and most recently [11C]MeS-IMPY. All these IMPYs have shown to rapidly cross the blood brain barrier (BBB) and bind to their target with high affinity.
We conducted a structure and activity relationship study of over 30 IMPYs on the following proteins: tau-PHFs, Ab oligomers, Ab1-42 fibrils, g-secretases, a-synuclein and prion protein. Furthermore, we performed histochemical staining of plaques and tangles on human post mortem brain slices. This unique, parallel approach, allows for a comparative study of the ligands binding mode onto each protein – providing crucial information for developing a selective ligand for diagnosis and treatment.
Project 2:
Today, a postmortem diagnosis of Alzheimer’s disease is relied on the accurate quantification of its hallmark proteins – amyloids and tangles. On staining with hematoxylin and eosin, amyloid stains no differently from many other proteins. The amyloid proteins’ specific underlying organization affords various staining procedures for their detection, for example with Congo red, silver, thioflavin-S, and immunohistochemical staining techniques.
Common drawbacks of these methods are nonspecific binding, stain variability, tissue damage, and the amount of time required for the procedure. We are developing several new histochemical dyes that can reveal either amyloids or tangles selectively under a fluorescence microscope. Furthermore, these molecules follow Lipinski’s rule of 5 thus have high probability to cross the blood brain barrier, allowing for both in vitro and in vivo monitoring of amyloid and tangles.