Refine
Year of publication
Document Type
- Doctoral Thesis (21)
Has Fulltext
- yes (21)
Is part of the Bibliography
- no (21)
Institute
- Biowissenschaften (13)
- Medizin (8)
In der Neurobiologie nimmt die Untersuchung der Großhirnrinde (Neokortex) eine gewisse Sonderstellung ein, weil das Verständnis dieser hierarchisch übergeordneten Region für die Analyse bzw. Rekonstruktion der Hirnfunktionen insgesamt von entscheidender Bedeutung ist.
Dabei macht Folgendes die bedeutungsvolle Stellung des Neokortex aus: seine späte stammesgeschichtliche und ontogenetische Entwicklung, welche bei mehreren Säugetiergruppen mit einer ungewöhnlichen Massenzunahme und Plastizität verbunden ist und letzten Endes auch Raum für Individualität und Intelligenz schafft. Dabei kommt es speziell bei Primaten inklusive des Menschen zu einer zunehmenden Diversifizierung in Areale, welche primär den Sinnessystemen (Sehen, Hören, Tastsinn) sowie der Motorik zugeordnet sind. Mit steigender "Evolutionshöhe" der Säugetiere treten aber auch hierarchisch übergeordnete sekundäre, tertiäre und weitere Areale auf, welche zunehmend der Assoziation bzw. Integration von Sinnessystemen gewidmet sind. All diese Areale steuern die Interaktion des Individuums mit seiner Umwelt, d.h. sie formulieren anhand des aus der Peripherie eingehenden afferenten Inputs eine biologisch sinnvolle (motorische) Reiz-Antwort und ermöglichen in ihrer Gesamtheit (vor allem beim Menschen) auch kognitive Prozesse, so z.B. multisensorisches und assoziatives Denken, aber auch Antrieb, Planung, Erinnerung und ein hochkompliziertes Sozialverhalten.
Die Zielsetzung der vorliegenden Arbeit besteht darin, bei verschiedenen, teilweise extrem unterschiedlichen Säugetiergruppen über die vergleichende Morphologie der primären Neokortex-Areale zu einem besseren Verständnis grundsätzlicher neokortikaler Funktionsprinzipien (Input, intrinsische Verschaltung, Output) beizutragen. Die Einbeziehung phylogenetischer Aspekte kann dabei helfen, die kortikalen Spezifika der jeweiligen Säugetiere auf ihren Anpassungswert hin kritisch zu überprüfen.
Im Detail werden die vier primären Rindenfelder des auditorischen [A1], motorischen [M1], somatosensiblen [S1] und visuellen Systems [V1]) bei Spezies aus unterschiedlichen Ordnungen wie den Primaten (Mensch, Gorilla), Raubtieren (Hund), Paarhufer (Artiodactyla: Schwein, Schaf) sowie der Wale und Delphine (Zahnwale oder Odontoceti; u.a. Großer Tümmler, Schweinswal) anhand einer ganzen Palette von qualitativen und quantitativen Methoden konsequent miteinander
verglichen. Als eine solide Basis dient hier die allgemeine Zytoarchitektonik (Nissl- und teilweise Golgi-Färbung), welche durch immunhistochemische Marker (Calbindin, Calretinin, Parvalbumin und Neurofilament) um eine funktionell-neurobiologische Ebene erweitert wird. Die neben den Primaten im Fokus stehenden Delphine, welche sich durch eine erstaunliche Uniformität ihrer Großhirnrinde auszeichnen, werden mittels der “design-basierten“ Stereologie zusätzlich auf die Neuronendichte der kortikalen Areale bzw. ihrer Rindenschichten hin untersucht. Dabei wurden anhand phylogenetischer und evolutionsbiologischer Überlegungen jeweils die Rindenschichten III-V als "Schlüsselregion" ausgewählt, um über die Berechnung von Neuronendichten innerhalb dieser Schichten III und V mehr über die funktionellen Implikationen dieses eigentümlichen Neokortex herauszufinden.
Insgesamt zeigt sich, dass der Neokortex im Laufe der Evolution wohl gerade bei den landlebenden Primaten besonders stark diversifiziert worden ist: ihre vier primären Rindenfelder unterscheiden sich im Vergleich mit anderen Säugetieren besonders deutlich: hinsichtlich der Rindengliederung, der Ausstattung mit Neuronentypen sowie der intrinsischen Verschaltung erreicht dieser Kortex ein Höchstmaß an Komplexität. Besonders deutlich wird dies in den granulären Arealen, welche besonders viele Körnerzellen aufweisen (vor allem in der inneren Körnerzellschicht, Lamina IV).
Demgegenüber finden sich bei den holaquatischen Delphinen stark abweichende Verhältnisse. Ihre Großhirnrinde erscheint nicht nur allgemein recht einheitlich bzw. monoton, sondern auch in ihrer intrinsischen Funktionsweise stark abgeleitet: hier zeigt sich ein genereller Trend zur Entwicklung einer uniformen Rinde auf gänzlich pyramidal-agranulärer Basis, welche durch das Fehlen einer deutlich erkennbaren Schicht IV gekennzeichnet ist.
Bei einem Vergleich der bearbeiteten Säugetiergruppen ergibt sich ein neokortikales Kontinuum zwischen der granulären und der pyramidalen Bauweise zweier herausgehobener “Modellsäuger“, welche einander gewissermaßen als morphologische und funktionelle Extreme gegenüberstehen: mit den Primaten als dem einen (granulären) Endpunkt und den Delphinen als dem anderen (pyramidalen) Endpunkt sowie dazwischen vermittelnden Übergangsformen.
Die Hirnrinde des terrestrischen Karnivoren (Hund) zeigt dabei zuweilen Charakteristika, welche ihn eher in die Nähe der Primaten rücken. Bei den
terrestrischen Paarhufern (Schaf, Schwein) finden sich dagegen manche Kortex-Merkmale, welche an die Situation bei Delphinen erinnern.
Von besonderer Bedeutung ist hier nun die Feststellung, dass wohl gerade die sekundäre Anpassung der Zahnwale (Delphine) an eine ausschließlich aquatische Lebensweise für die Ausbildung ihres ureigenen Typus von Kortex entscheidend gewesen sein dürfte. In diesem Zusammenhang werden die physikalischen Eigenschaften von Wasser die Rahmenbedingungen für evolutionäre Abwandlungen innerhalb des Gehirns als Ganzem vorgegeben haben. Interessanterweise werden neben den höchst- enzephalisierten Vertretern unter den Primaten (Hominidae; Mensch und Menschenaffen) auch den Zahnwalen (Delphinen) von manchen Neurobiologen herausragende kognitive und intellektuelle Fähigkeiten zugeschrieben - trotz der diametral unterschiedlichen Organisation ihres Kortex. Ob und inwieweit dies zutrifft, bleibt weiteren Untersuchungen vorbehalten.
Die Ergebnisse der vorliegenden Arbeit verlangen für die Zukunft nach weiteren gleichartigen Untersuchungen der nicht-primären, also hierarchisch übergeordneten neokortikalen Assoziationsareale im Hinblick auf funktionelle und evolutions-biologische Implikationen.
Die überwältigende Expansion des Neokortex und die daran gekoppelte starke Zunahme der neurobiologischen Kapazität des Gehirns insgesamt dürfte nicht nur bei den Primaten, sondern (in bescheidenerem Umfang) auch bei Delphinen zur Entstehung von kortikalen "Freiräumen" geführt haben. Beim Menschen handelt es sich bei diesem Zuwachs an Hirnmasse und Hirnkapazität vermutlich um das Substrat für die Entstehung der phylogenetisch jüngsten und kognitiv höchstabgeleiteten Merkmale, nämlich Sprache, Persönlichkeit und Vernunft.
The present study approached two related but conceptually different questions of EV biology in cancer. In both approaches, tailored variants of the Cre LoxP system were utilized. First, in the context of intradermal and intracranial tumours, it was examined which cells in the tumour microenvironment (TME) take up tumour derived
EVs and what effects EV uptake has on recipient cells. Secondly, in the context of glioma, peripheral macrophages (MF) were directly traced to the brain and
separated from brain resident microglia (MG). Furthermore, EV signalling between these entities was analysed.
Regarding the first approach, multidirectional transfer of functional Cre recombinase RNA in intradermal and intracranial mouse tumour models was observed. In spite of robust recombination rates in all tumour models, the total number of EV-uptaking cells is around three times higher than the total number of recombined cells, suggesting that interactions of cells and EVs which contain CremRNA does not necessarily lead to marker gene expression. Subsequent studies can build up on this established system and isolate and characterise EV-uptaking cells to identify geno- and phenotypical changes induced by EV uptake.
The second, conceptionally different aspect that was investigated in this study is the distinction and tracing of peripheral MF to the brain and their distinction from
brain resident MG in glioma. Glioblastoma multiforme (GBM) is the most common and the most malignant brain tumour. The average patient survival of 15 months
past diagnosis did not change much during the last decades, which stresses the need for new therapies. GBM location in the immune privileged brain, its characteristically highly immune suppressive TME and its highly invasive growth
makes this disease so difficult to treat. Immune therapies, which in general show good results in other types of cancer, are not effective in GBM. To a great extent, this can be ascribed to the lack of understanding of MG and MF function in GBM and their roles in tumour progression.
Cellular communication is a concept that can be explained as the transfer of signals or material (such as cytokines, ions, small molecules) between cells from the same or different type, across either short or long distances. Once this signal or material is received, it will, as a rule, promote a functional effect. Several routes, involved in this transfer, are well described and are of global importance for organ/tissue communication in an organism.
The brain interacts dynamically with the immune system, and the main route known to mediate this communication, is via the release of cytokines (by peripheral blood cells), which can then activate certain brain cell types, such as microglia, directly, or activate the vagus nerve transferring signals to neuronal populations in the brain. The communication between these two systems plays a key role in the pathophysiology of neurodegenerative diseases, and the mechanisms involved in this interaction are of central importance for understanding disease initiation and progression and search for therapeutic models.
The Momma lab previously addressed the mechanisms of interaction between the peripheral immune system and the brain by investigating cellular fusion of haematopoietic cells with neurons after inflammation. They addressed the question of whether this phenomenon also occurs under non-invasive conditions. To approach this problem, a genetic tracing model that relies on the Cre-Lox recombination system was used. Transgenic mice expressing Cre recombinase specifically in the haematopoietic lineage were crossed into a Cre-reporter background, thus all haematopoietic cells irreversibly express the reporter marker-gene EYFP. Using this model, EYFP was detected in non-haematopoietic tissues, suggesting the existence of a communication mechanism never described before. As cells containing two nuclei were never detected, fusion as a mechanism was excluded, suggesting that Cre reaches non-haematopoietic cells via a different signalling pathway. The Momma lab investigated whether the transfer of material through extracellular vesicles (EVs) could be behind this periphery-to-brain communication. Using the genetic mouse model, they were able to trace the transfer of Cre RNA via EVs between cells in vivo, generating the first in vivo evidence of functional RNA transfer by EVs between blood and brain.
The last decade has witnessed a rapid expansion of the field of EVs. Initially considered as waste disposal material, recent evidence has challenged this view. EVs are currently considered as a widespread intercellular communication system that can transport and transfer all types of biomolecules, from nucleic acids to lipids and proteins. However, several important questions are still under investigation. One of them is whether EVs are involved in brain pathophysiology, as inflammation plays an important role in onset and progression of neurodegenerative diseases and is well described in Parkinson Disease (PD). Based on preliminary data in a mouse, peripherally injected with a low dose of Lipopolysaccharide (LPS, an endotoxin found in the outer-membrane of Gram-negative bacteria, which causes an immune response), neurons and other cell population in the brain take up EVs from the periphery. Particularly, dopaminergic neurons from Substantia Nigra and Ventral Tegmental Area have been shown to receive functional RNA, transported through EVs, which can lead up to 20% of recombination. Furthermore, different neuronal populations from Hippocampus, Cortex and Cerebellum exhibit recombination, indicating a widespread signalling from blood to the brain. Therefore, the goal of my PhD thesis was to investigate the mechanisms of this transfer and the triggers that lead to EV uptake by neural cells in vivo both in pathological and physiological conditions.
In this project, the extent and function of EV-mediated signalling from blood to brain is explored in the context of peripheral inflammation and neurodegenerative diseases. Firstly, EVs isolated from WT mice were further characterized using size-exclusion chromatography (SEC), Western Blot (WB) and electron microscopy in order to extend the knowledge from previous work done in the Momma lab. Secondly, to expand on the biological relevance of the fact that inflammation is correlated with an increase in EV uptake, different approaches using the genetic murine tracing model were used. Recombination events from haematopoietic cells to the brain have been followed after peripheral injection of LPS. Peripheral inflammation caused by LPS injection led to widespread recombination events in the brain, specifically in microglia and neurons, including dopaminergic (DA) neurons. In contrast, astrocytes, oligodendrocytes and endothelial cells were never or very rarely recombined. Additionally, peripheral LPS injection in a murine model, where Cre is expressed only in erythrocytes, led to recombination events only in microglia, suggesting that the type of EV-secreting cell plays a role in the targeting of EVs to a specific cell population.
EGFL7 regulates adult neural stem cell maintenance and differentiation by inhibition of Notch1
(2010)
In neurobiology the preexisting dogma on the unchangeability of the adult mammalian brain and its inability to give rise to new neurons has been challenged since the early nineties. Generally, it is now accepted that neurogenesis persists in adults. Progress in developmental and stem cell biology in recent years led to an increasing interest in regeneration-based treatment strategies for damaged tissue of the central nervous system. Thus, the enhancement of the endogenous potential of the brain to repair itself is potentially a feasible therapeutic strategy to treat various types of brain damage. Therefore, it is of great interest to understand the molecular mechanism that regulate adult neurogenesis. One of the prominent pathways suggested to be involved here is the Notch signaling cascade. Previously, it has been shown that various components of Notch signaling are expressed in the stem cell niche of the adult subventricular zone (SVZ) in vivo. Interestingly, a recent study demonstrated that the self-renewal potential of adult neural stem cells (NSCs) isolated from the SVZ depend on Notch signaling in vitro.
Recently, we identified a novel non-canonical Notch ligand termed epidermal growth factor-like domain 7 (EGFL7), which was originally described as a protein secreted by endothelial cells and functionally implicated in cellular responses of the vascular system such as cell migration and blood vessel formation. We were able to show that secreted EGFL7 binds to a region in Notch that is involved in ligand-mediated receptor activation, thus acting as an antagonist of Notch signaling.
The present study identifies neurons of the human and murine brain as a novel source of EGFL7, which suggests functions of EGFL7 in the neural system. Expression analyses by quantitative RT-PCR (qRT-PCR) revealed EGFL7 is down regulated in the adult SVZ, which suggests that endogenous EGFL7 may act as a Notch modulator of NSCs. We assessed the expression of Notch pathway components in adult NSCs isolated from the SVZ of adult mice and demonstrated that inhibition of Notch activity by the γ-secretase inhibitor DAPT reduced the self-renewal potential of NSCs. Accordingly, adenoviral-mediated expression of EGFL7 in vitro decreased Notch-specific signaling and reduced proliferation and self-renewal of NSCs. Conversely, activation of Notch by a constitutive active form of Notch (NICD) rescued the EGFL7-mediated reduction of NSC self-renewal verifying that this effect was directly linked to Notch signaling. Congruent to the reduced proliferation rate measured in vitro, induced expression of EGFL7 in vivo significantly reduced the number of Ki-67 positive cells within the SVZ upon cerebroventricular injection of EGFL7 adenovirus.
Expression analyses in the developing brain showed single EGFL7-positive cells within the marginal zone of the neocortex as measured by in situ hybridization. These cells might be Cajal-Retzius cells, specialized neurons, which specifically express Reelin, which is a protein of the extracellular matrix known to control neuronal migration and differentiation. Interstingly, we could show that Reelin and EGFL7 are expressed in a subtype of neurons of the adult mouse cortex. This implied an interaction of both proteins and was verified by co-immunoprecipitation assays, suggesting an additional role for EGFL7 in neuronal maintenance. QRT-PCR based expression analyses in vitro comparing differentiated and non-differentiated NSCs displayed an increase in EGFL7 expression during the differentiation process, which was paralled by reduced Notch signaling. NSCs differentiated on coverslips coated with EGFL7 differentiated into all three cell types - neurons, oligodendrocytes and astrocytes. EGFL7 favored the formation of neurons as compared to control comparable to the effect of the Notch-inhibitor DAPT. Furthermore, additional oligodendrocytes were formed. These cells displayed a mature morphology with distinct sprouts and branches in contrast to the small and round oligodendrocytes that formed on control coverslips, which resembled us of precursor cells. Neurons and oligodendrocytes were formed at the expense of astrocytes. Congruently to the effect observed in vitro, adenoviral-based expression of EGFL7 in the SVZ yielded a slight induction of neuronal differentiation in vivo. Taken together, these results show for the first time a previously unrecognized role for EGFL7 in the brain by modulation of the Notch pathway in adult NSCs, which changes the proliferation and differentiation potential of adult NSCs in vitro and in vivo.
Stem cells are often referred to as potential candidates for the treatment of different pathologies. Their ability to differentiate into various tissue specific cell types offers the possibility to engineer cell systems or organs for replacement. One of the main questions in stem cell biology is how stemness properties are regulated and to what extend this regulation is intrinsic or conveyed by the direct microenvironment (‘niche’). In order to elucidate such regulatory processes, it is informative to analyze processes or molecules that are shared between different stem cell populations.
One such molecule that is expressed on a wide range of different embryonic and adult as well as tumor stem cells is the ABC transporter Abcg2. ABC transporters in general are transmembrane proteins that actively extrude endo- and exotoxins as well as xenobiotics, thereby protecting cells and organs. Additionally, ABC transporters are responsible for drug resistance in many cancers. A well-described characteristic of stem cells expressing Abcg2 is the formation of the ‘side population’ (SP) phenotype: An active Abcg2 transporter mediates the efflux of a particular fluorescent dye that is taken up by all cells, thus leading to a less brightly stained population. This phenomenon is widely used to characterize and isolate the most primitive stem cell subpopulation from embryonic and adult tissues, including tumors. Besides its role as toxin transporter little is known about the function of Abcg2 in stem cells. This is mainly due to the fact that its physiological substrate in stem cells remains unknown. The identification of such substrates is therefore of high interest because it would directly link the activity of ABC transporters to regulatory mechanisms in stem cell biology.
In the present study we wanted to test the hypothesis that the sphingolipid ceramide is a physiological substrate of the ABC transporter Abcg2. Sphingolipids are potent second messengers and are known to have regulatory functions in stem cells. In particular, the sphingolipid ceramide is described as a mediator of controlled cell death and inducer of differentiation. It is suggested that stem cells need to keep their intracellular ceramide content at low levels in order to prevent apoptosis or differentiation. We propose that Abcg2 and ceramide interact and that this interaction leads to changes in the absolute or relative amounts of ceramide. This in turn influences basic stem cell functions such as self renewal and differentiation.
We show that Abcg2 prevents cells from accumulating fluorescence labeled ceramide. Furthermore, exogenously applied ceramides inhibit the transport activity of Abcg2, measured by a decrease of the side population phenotype. This inhibitory effect is consistent with a competitive inhibition mechanism. Additionally, we show that active Abcg2 can increase the ceramide concentration in cell culture supernatant. Finally we demonstrate that Abcg2 protects from ceramide induced cytotoxicity in human cell lines. In summary, these in vitro results strongly suggest that Abcg2 has the ability to regulate ceramide levels.
Murine hematopoietic stem cells (HSCs) are the best characterized adult stem cell system so far. By using 7-colour fluorescence-activated cell sorting (FACS) we established the purification of the most primitive HSCs, reflected by their high engraftment capability when transplanted to lethally irradiated mice. By using this sorted cell populations it was in addition possible to establish a system to reproducibly manipulate HSCs ex vivo. This experimental system will serve in further elucidating the physiological consequences of Abcg2 mediated changes in ceramide levels on stem cells in vivo.
Taken together, this study shows that Abcg2 has the ability to regulate ceramide levels in cells. This in turn can lead to cellular protection from ceramide induced apoptosis. Additionally, the experimental techniques to further analyze the role of Abcg2 and ceramide in the most primitive hematopoietic stem cells were successfully established, enabling more detailed analysis in the future.
Glioblastoma is the most common and most aggressive type of brain tumor in adults. In contrast to epithelial cancers, glioblastomas do not metastasize. While the major treatment challenge in epithelial cancers is not the primary tumor but metastasis, glioblastoma patients die of the primary tumor.
However, there is a common theme which underlies the malignant properties of progressed epithelial cancers and glioblastoma: invasion from the primary tumor into the surrounding tissue. In the case of epithelial cancers this is the first and necessary step to metastasis, whereas invasion leads inevitably to tumor recurrence after resection in the case of glioblastoma, causing it to be incurable.
A cellular program which has been described in detail to promote the invasive phenotype in epithelial tumors, is the epithelial-mesenchymal-transition (EMT). Differentiated neural cells are not epithelial, thus, strictly speaking, EMT does not occur in glioblastoma. However, the traits acquired in the process of EMT, especially invasiveness and stemness, are highly relevant to glioblastoma. One of the key transcription factors known to induce EMT in epithelial cancers is ZEB1, which has been described only marginally in the central nervous system so far. Here, I investigate the expression and function of ZEB1 in glioblastoma and during human fetal neural development.
ZEB1 mRNA was significantly upregulated in all histological types of glioma, including glioblastoma, when compared to normal brain. There was no correlation between ZEB1 mRNA levels and tumor grade. Immunohistochemical staining of glioma samples demonstrated that ZEB1 was highly expressed in the great majority of tumor cells. In the developing human brain, intense staining for ZEB1 could be observed in the ventricular and subventricular zone, where stem- and progenitor cells reside. ZEB1 positive cells included cells stained with stem- and progenitor markers like PAX6, GFAP and Nestin. In contrast, ZEB1 was never found in early neuronal cells as identified by TUBB3 staining.
To gain insight into ZEB1 function I generated a human fetal neural stem cell line and a glioblastoma cell line with ZEB1 knockdown, which were compared with their respective control cell lines. First, I found that ZEB1 does not regulate the micro RNA 200 family in either cell line, which has been described as an essential ZEB1 target in epithelial cancers. Second, regulated target genes were identified with a genome wide microarray. The third approach was to directly identify genomic binding sites of ZEB1 by chromatin immunoprecipitation sequencing (ChIP-seq). All three approaches showed that the ZEB1 transcriptional program is surprisingly similar in the neural stem cell line and the glioblastoma cell line. In contrast, it bears only little resemblance to the program described in epithelial cancers.
The most interesting, previously unrecognized ZEB1 target gene identified in this study is integrin b1. It was regulated after ZEB1 knockdown detected by microarray analysis, and has a ZEB1 binding site in its promoter region detected by ChIP-seq. Finally, I addressed the question whether ZEB1 influences tumor growth and invasiveness in a glioblastoma model. After intracranial xenotransplantation in mice, ZEB1 knockdown glioblastoma cells formed significantly smaller and less invasive tumors than control glioblastoma cells.
This study demonstrates that ZEB1 is widely expressed in glioma and relevant for glioblastoma growth and invasion. In contrast to what is known about ZEB1 function in epithelial cancers, ZEB1 is not associated with glioma progression, but instead seems to be an early and necessary event in tumorigenesis. Also with regard to ZEB1 target genes, ZEB1 functions differently in glioblastoma than in epithelial cancers. The two most important ZEB1 targets in epithelial cancers are E-cadherin and the miR-200 family members. Both are not relevant to ZEB1 function in glioblastoma. Interestingly, while the ZEB1 transcriptional program is different from the one described in epithelial cancers, it is highly similar in glioblastoma cells and fetal neural stem cells. This suggests that an embryonic pathway restricted to stem- and progenitor cells during development is reactivated in glioblastoma.
Previously known ZEB1 target genes were tissue specific and therefore seemed unlikely to mediate ZEB1 function in the central nervous system. However, the newly identified ZEB1 target gene integrin b1 is well known to play pivotal roles in both glioblastoma tumorigenesis and invasion as well as in neural stem cells. Additionally, integrin b1 is widely expressed and seems a likely ZEB1 target in other organs than the brain.
Taken together, I demonstrate that ZEB1 is a new regulator of glioblastoma growth and invasion. The transcriptional program of ZEB1 differs from the one in epithelial cancers but is strikingly similar to the one in neural stem cells. The newly identified ZEB1 target gene integrin b1 is likely to mediate crucial ZEB1 functios. Thus, this study identifies ZEB1 as a yet unrecognized player in glioblastoma and neural development. Furthermore, it sets the stage for more research which will help to deepen our understanding of ZEB1 function in the central nervous system and beyond.
Einleitung In ihrer klinischen Symptomatik lassen sich der ischämische Schlaganfall (IS) und die intrazerebrale Blutung (ICH) nicht sicher unterscheiden. Hinsichtlich der Akuttherapie, die eine möglichst schnelle Wiederherstellung der zerebralen Sauerstoffversorgung („time is brain“) zum Ziel hat, ist diese Differenzierung jedoch essentiell. Das Ziel der vorliegenden Arbeit ist der Nachweis serologischer Biomarker in der Frühphase des Schlaganfalls zur Differenzierung zwischen IS und ICH. Hypothesen-gestützt wurden aufgrund pathophysiologischer Überlegungen hierfür die ZNS-spezifischen Proteine GFAP, UCH-L1, pNf-H, MBP und Tau untersucht. In einem hypothesenfreien Ansatz wurden Serumproben von Schlaganfallpatienten massenspektrometrisch analysiert.
Material und Methoden Die Patientenrekrutierung für die vorliegende Arbeit erfolgte im Rahmen der prospektiven, multizentrischen BE FAST II-Studie zur Evaluation von GFAP als Biomarker der akuten ICH. Von Mai 2012 bis April 2014 wurden Serumproben von Patienten mit akuter Schlaganfallsymptomatik in der Klinik für Neurologie der Goethe-Universität Frankfurt am Main gesammelt. Mittels kommerziell verfügbaren ELISA-Kits wurden die Serumkonzentrationen der Proteine UCH-L1, pNf-H, MBP und Tau bestimmt. Die Analyse der GFAP-Serumkonzentration erfolgte durch Roche Diagnostics mit Hilfe des Elecsys®-Systems, einem Elektrochemiluminiszenz-Immunoassay. Für die Massenspektrometrie wurden Serumproben aus der BE FAST-I-Studie, die von Ende des Jahres 2010 bis Anfang des Jahres 2011 asserviert wurden, eingesetzt. Die massenspektrometrischen Untersuchungen wurden in der Abteilung „Funktionelle Proteomics“ im Fachbereich Medizin der Goethe-Universität Frankfurt am Main durchgeführt.
Ergebnisse Tau und MBP ließen sich in den meisten Serumproben nicht nachweisen. In der pNf-H-Messung lag die Konzentration bei 27 von 35 Patienten oberhalb der Nachweisgrenze, wobei sich kein signifikanter Unterschied zwischen IS und ICH ergab (p = 0,69). UCH-L1 ließ sich bei 28 von 29 Patienten nachweisen. In der IS-Gruppe war eine signifikant (p = 0,005) höhere UCH-L1-Konzentration nachweisbar (Median 5,71 ng/ml) als in der ICH-Gruppe (Median 2,37 ng/ml). GFAP war bei allen 45 Patienten nachweisbar mit signifikant (p < 0,00005) höherer Konzentration in der ICH-Gruppe (Median 2,87 ng/ml) verglichen mit der IS-Gruppe (Median 0,01 ng/ml). Zudem fand sich eine positive Korrelation der UCH-L1-Werte in der IS-Gruppe mit dem Patientenalter (r = 0,62, p = 0,01), sowie eine positive Korrelation der GFAP-Werte in der ICH-Gruppe mit dem Patientenalter (r = 0,54, p = 0,03), dem NIHSS-Wert (r = 0,69, p = 0,04) und mit dem ICB-Volumen (r = 0,60, p = 0,01). In der massenspektrometrischen Analyse ließ sich eine Top Liste aus 22 Proteinen erstellen, die jeweils signifikante Unterschiede zwischen IS und ICH aufweisen.
Diskussion Die Rolle des Ubiquitin-Proteasom-System (UPS) und insbesondere von UCH-L1 beim IS ist bislang noch nicht abschließend geklärt. Nach einer zerebralen Ischämie ist jedoch eine Upregulation von UCH-L1 beschrieben, u.a. durch eine verstärkte UPS-Aktivität durch Aggregate fehlgefalteter Proteine. Daneben reagieren Neurone sensibler auf eine Hypoxie als Gliazellen mit einer dominierenden Freisetzung neuronaler Proteine wie UCH-L1. Bei ICH kommt es dagegen eher zu einer unspezifischen Destruktion des Hirngewebes mit vorwiegender glialer Schädigung und rascher Freisetzung glialer Proteine wie GFAP. Mit UCH-L1 und GFAP konnten zwei Proteine als erfolgsversprechende Kandidaten zur Differenzierung zwischen IS und ICH in der Frühphase identifiziert werden. Zur weiteren Validierung sind Untersuchungen an einer großen Population notwendig, die auch kleinere Infarkte und Hirnblutungen einschließt. Auch der Einfluss epidemiologischer und klinischer Faktoren wie z.B. dem Patientenalter muss weiter evaluiert werden.
Die mittels Massenspektrometrie erstellte Top Liste aus 22 Proteinen enthält vielversprechende Biomarker-Kandidaten, die signifikante Unterschiede zwischen IS und ICH aufweisen und ebenso an einem großen Patientenkollektiv weiter untersucht werden müssen.
The canonical Wnt/β-catenin and the Shh pathway as well as the Notch signaling cascade
are key regulators in stem cell biology and are independently associated with the development
of cancer. Despite the knowledge of a balanced signaling for cellular maintenance, the
fundamental biochemical mechanisms of crosstalk are still poorly understood. This study
demonstrates that the outcome of interaction between Wnt and Shh is cell type specific. A
combined inhibitory mechanism of the Shh and Notch2/Jagged2 pathways on dominant
active β-catenin signaling in the adult tongue epithelium keeps Wnt/β-catenin signaling
restricted to physiological tolerable levels. In the opposite crosstalk the activation of
Wnt/β-catenin signaling in medulloblastoma (MB) of the Shh subtype, in turn inhibits the Hh
pathway.
The inhibitory mechanism of Shh and Notch2/Jagged2 on Wnt/β-catenin signaling is
independent of the degradation complex of β-catenin and takes place inside the nucleus.
Furthermore, the negative feedback on Wnt/β-catenin signaling by the Shh pathway relies
on transcriptional activity of Gli1/2A. Inhibition of Gli1/2A with the specific inhibitor GANT61
abrogated the negative impact of Shh on β-catenin signaling in vitro. Although the negative
feedback loop of Shh is still functional in human SCC25 cells, the inhibitory effect of
Notch2/Jagged2 is lost and contributes to the cancerogenic phenotype of these cells. In the
inverse situation, the activation of β−catenin signaling has a negative feedback on
constantly active Shh signaling and significantly inhibits the Hh pathway. This was shown in
Ptch+/- and Math1-Cre:SmoM2Fl/+ MB tumor spheres in vitro, in which inhibition of sphere
formation and growth was observed and Hh target gene transcription was down-regulated.
This demonstrates for the first time that the activation of canonical Wnt/β-catenin signaling
in primary MB cells with a Hh pathway over-activation has a negative effect on the growth of
these cells in vitro.
In summary the results show that crosstalk of Wnt/β-catenin and Shh signaling has context
specific outcome on pathway activity. Elucidation of the molecular interactions will improve
our understanding of Wnt and Hh associated tumors and contribute to the development of
new therapeutic strategies.
Blood vessel formation is a well orchestrated process where multiple components including different cells types, growth factors as well as extracellular matrix proteins act in synergistic and highly regulated manner to support the growth of new blood vessels. During embryonic development this process is marked as vasculogenesis and entails the differentiation of mesodermal cells into angioblasts and their subsequent fusion into a primitive vascular plexus. Angiogenesis, in contrast, describes the formation of new vessels from the pre-existing vasculature and it occurs in the embryo during remodeling of the primitive plexus into a mature vascular network. Furthermore, in the adult, angiogenic processes play a role in various physiological and pathological conditions. Angiogenesis is governed by a set of factors and molecular mechanisms whose identification has been a major focus of cardiovascular research for the past several decades. Most recently, Epidermal growth factor-like domain 7 (EGFL7) has been described as a novel molecular player in this context. This secreted protein is produced by endothelial cells and has been implicated in vessel development. Studies performed in zebrafish revealed an important role for EGFL7 in lumen formation during vasculogenesis although the underlying molecular mechanism has not been elucidated yet. In contrast, the investigation of EGFL7’s functions during angiogenic sprouting has faced several challenges and the role of EGFL7 in angiogenesis remained elusive. The purpose of this thesis was to identify the functions of EGFL7 during angiogenic mode of vessel formation in a systematic fashion using numerous in vitro as well as in vivo approaches.
Previously it has been suggested that EGFL7 might associate with the extracellular matrix from where it could exert its effects. Indeed, we could show that EGFL7 accumulates on the outer surface of endothelial cells in vivo by demonstrating its co-localization with collagen IV, a major constituent of the basal lamina. Furthermore, after its secretion to the extracellular matrix (ECM), EGFL7 seemed to interact with some components of the extracellular matrix including fibronectin and vitronectin, but not collagens and laminin.
A major group of receptors that mediate the interaction between the cells and the ECM are integrin receptors. Our co-immunoprecipitation studies revealed that EGFL7 associated with integrin αvβ3 which is highly expressed in endothelial cells and known to be important for vessel growth. Importantly, this EGFL7-αvβ3 integrin interaction was dependent on Arg-Gly-Asp (RGD) motif present within the second EGF-like domain of EGFL7 protein. Adhesion assays performed with human umbilical vein endothelial cells (HUVEC) revealed that EGFL7 promoted endothelial cell adhesion compared to BSA used as a negative control, however, adhesion seemed to be less efficient as compared to bona fide ECM proteins such as fibronectin and vitronectin. In addition, cultivation of endothelial cells on EGFL7 was characterized by the absence of mature focal adhesions and stress fibers, but was paralleled by increased phosphorylation of kinases typical for integrin activation signaling cascade such as FAK, Src and Akt. This led us to the hypothesis that EGFL7 creates an environment that supports a motile phenotype of endothelial cells by serving as a modulator of existing interactions between the cells and the surrounding matrix. Indeed, EGFL7 increased random migration of HUVEC on fibronectin in an αvβ3 integrin dependent manner as shown using a live cell imaging platform. Most importantly, this was paralleled by a decrease in endothelial cell adhesion to fibronectin which is consistent with previous reports on secreted proteins that support a medium strength of adhesion and such promote cellular migration. To assess the overall effect of EGFL7 on the process of blood formation several in vitro and in vivo approaches were employed. First, the addition of EGFL7 to Matrigel injected subcutaneously into mice significantly increased the invasion of endothelial cells into the plugs. Second, a spheroid-based sprouting assay in three-dimensional collagen matrix clearly demonstrated the ability of EGFL7 to support angiogenic sprouting in an integrin dependent manner. This is consistent with the observed effects of EGFL7 on endothelial cell migration. Third, using in vivo assays such as the chick chorioallantoic membrane (CAM) assay as well as a zebrafish model system we were able to validate the importance of the EGFL7-integrin interaction for the process of angiogenesis in vivo. Taken together, I identified some of the major cellular functions EGFL7 modulates during angiogenesis. In addition, with integrin αvβ3 I unraveled a novel interaction partner of EGFL7 that delivers a mechanistical explanation for EGFL7’s effects on blood vessel formation. Most importantly, data presented in this PhD thesis contribute substantially to the existing literature on EGFL7 unambiguously assigning a role for this protein in the process of angiogenesis.
The canonical Wnt pathway, also known as Wnt/β-‐catenin pathway, comprises a network of proteins which control diverse developmental and adult processes in all metazoan organisms. The binding of canonical Wnt ligands to a cell surface receptor complex, consisting of frizzled family members and low density lipoprotein receptor-‐ related protein 5 or 6 co‐receptors, triggers a signaling cascade which results in a β-catenin-‐mediated transcriptional activation of different target genes, implicated in cellular proliferation, apoptosis, migration and differentiation. A couple of years ago, several groups including us, iden2fied transient activation of the canonical Wnt-pathway in endothelial cells (ECs) of the developing central nervous system (CNS). In this context, Wnt/β-‐catenin signaling could be demonstrated to be crucial for brain angio genesis as well as for the establishment of the blood-brain barrier (BBB) phenotype in the newly formed vessels.
Gliomas, in particular the glioblastoma (GBM), belong to the group of highly vascularized solid tumors which gain their vascularization due to an angiogenic switch occurring during tumor progression. Interestingly, nuclear localized β-‐catenin could be exclusively detected in the activated endothelium of induced rat gliomas and of human GBM, suggesting a so far unknown and not further characterized involvement of the canonical Wnt pathway in pathological angiogenesis. In order to systematically decipher the precise role of endothelial Wnt/β-‐catenin signaling in tumor angiogenesis, I established
murine GL261 glioma cell lines overexpressing either Wnt1 or Dickkopf (Dkk) 1 in a doxycycline-‐dependent manner, an activator and potent inhibitor of Wnt/β-‐catenin signaling, respectively. In subcutaneous and intracranial transplantations, tumor-derived Wnt1 reduced, while Dkk1 increased GL261 tumor growth without affecting in vitro proliferation, cell cycle or cell death of the established cell lines. Nowadays, it is well accepted that solid tumors are dependent on vascular support allowing them to grow beyond a certain size. In my work I could show that tumor-‐derived Wnt1 targets the tumor vasculature by increasing endothelial Wnt/β-‐catenin signaling, which reduced tumor vessel density and resulted in a more quiescent tumor vasculature. Furthermore, Wnt1-‐expression mediated tight association of smooth muscle cells (SMCs) and pericytes to the tumor endothelium, a phenotype which is unusual for tumor vessels and a described hallmark of tumor vessel normalization. In contrast, inhibition of endothelial Wnt/β-‐catenin signaling by Dkk1 mediated an opposing effect, characterized by endothelial hyper-proliferation and a tumor vasculature with a rough basal lamina distribution and loosely anached mural cells, indicative of a strong angiogenic activity. The described vascular effects in Wnt1-expressing GL261 tumors could be verified by subcutaneous transplantations of a rat glioma cell line constitutively expressing Wnt1. Furthermore, an applied in vivo MatrigelTM plug assay uncovered the reduction in vessel density upon Wnt1 simulation to be tumor cell independent, suggesting an EC-‐autonomous effect. This hypothesis was confirmed by subcutaneous transplantations of parental GL261 cells into mice with genetically generated endothelial β-‐catenin gain-of-function (GOF). The derived GOF tumor from this experiment comprised a quiescent and normalized tumor vasculature and phenocopied the vascular effects observed in Wnt1-expressing tumors.
Our previous work provided evidence that Wnt/β-‐catenin signaling contributes to the BBB phenotype of the developing CNS through the transcriptional regulation of the tight junction protein claudin-‐3. Furthermore, the coverage of pericytes to brain vessels has been described to correlate with BBB integrity. In agreement with these publications, vessels of intracranial Wnt1-‐expressing GL261 tumors retained or regained barrier properties, indicated by a reduced leakage of the tracer Evans blue and endogenous mouse immunoglobulin G and increased junctional localiza2on of the tight junction proteins claudin-‐3, -‐5 and zonula occludens-‐1.
Overall, we detected sustained endothelial Wnt/β-‐catenin signaling to induce a quiescent and normalized tumor vascularization. Interestingly, the Notch signaling pathway has been shown to inhibit the angiogenic tip cell and to promote the quiescent stalk cell phenotype via its ligand Delta-like ligand 4 (Dll4) and the receptors Notch1 and 4. Mechanistically, my work demonstrated for the first time that overactivation of endothelial Wnt/β-‐catenin signaling reactivated expression of Dll4 in the tumor endothelium, which could be shown in vitro to increase Notch signaling and to favor a stalk cell-like gene signature. Furthermore, we uncovered the platelet-derived growth factor subunit B (pdgm) as a novel transcriptional target of Wnt/β-catenin signaling in ECs. Hence endothelial-‐derived PDGF-‐B is known to promote the recruitment of mural cells, the upregulation of this factor might explain the increased SMC/pericyte coverage observed in the tumor vasculature upon sustained endothelial Wnt/β-‐catenin signaling which additionally might promote a cycle of vascular normalization.
Taken together, my work reveals several vascular effects, being mediated by reinforced endothelial Wnt/β-‐catenin signaling during tumor angiogenesis. While a moderate level of canonical Wnt signaling, observed in vessels of human astrocytomas and murine control tumors, is considered to be associated with tumor angiogenesis, dominant activation of this pathway in ECs is shown to limit angiogenesis and to promote a quiescent and normalized tumor vasculature with increased barrier properties. Furthermore, my work discovers pdgm as a novel target of canonical Wnt signaling in ECs.
The work presented in this dissertation therefore not only uncovers the role of endothelial Wnt/β-‐catenin signaling in tumor angiogenesis but additionally reveals this pathway to be a novel modulator in pathological vessel development which might proof to be a valuable therapeutic target for anti-angiogenic and edema glioma therapy.