Network


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

Hotspot


Dive into the research topics where Andreas Androutsellis-Theotokis is active.

Publication


Featured researches published by Andreas Androutsellis-Theotokis.


Nature | 2006

Notch signalling regulates stem cell numbers in vitro and in vivo

Andreas Androutsellis-Theotokis; Ronen R. Leker; Frank Soldner; Daniel J. Hoeppner; Rea Ravin; Steve W. Poser; Maria Adele Rueger; Soo-Kyung Bae; Raja Kittappa; Ronald D. G. McKay

The hope of developing new transplantation therapies for degenerative diseases is limited by inefficient stem cell growth and immunological incompatibility with the host. Here we show that Notch receptor activation induces the expression of the specific target genes hairy and enhancer of split 3 (Hes3) and Sonic hedgehog (Shh) through rapid activation of cytoplasmic signals, including the serine/threonine kinase Akt, the transcription factor STAT3 and mammalian target of rapamycin, and thereby promotes the survival of neural stem cells. In both murine somatic and human embryonic stem cells, these positive signals are opposed by a control mechanism that involves the p38 mitogen-activated protein kinase. Transient administration of Notch ligands to the brain of adult rats increases the numbers of newly generated precursor cells and improves motor skills after ischaemic injury. These data indicate that stem cell expansion in vitro and in vivo, two central goals of regenerative medicine, may be achieved by Notch ligands through a pathway that is fundamental to development and cancer.


Stroke | 2007

Long-Lasting Regeneration After Ischemia in the Cerebral Cortex

Ronen R. Leker; Frank Soldner; Iván Velasco; Denise K. Gavin; Andreas Androutsellis-Theotokis; Ronald D. G. McKay

Background and Purpose— Because fibroblast growth factor 2 is a mitogen for central nervous system stem cells, we explored whether long-term fibroblast growth factor 2 delivery to the brain can improve functional outcome and induce cortical neurogenesis after ischemia. Methods— Rats underwent permanent distal middle cerebral artery occlusion resulting in an ischemic injury limited to the cortex. We used an adeno-associated virus transfection system to induce long-term fibroblast growth factor 2 expression and monitored behavioral and histological changes. Results— Treatment increased the number of proliferating cells and improved motor behavior. Neurogenesis continued throughout 90 days after the ischemia, and the occurrence of newly generated cells with characteristics of neural precursors and immature neurons was most evident 90 days after treatment. Conclusions— Focal cortical ischemia elicits an ongoing neurogenic response that can be enhanced with fibroblast growth factor 2 leading to improved functional outcome.


Proceedings of the National Academy of Sciences of the United States of America | 2008

The mammary microenvironment alters the differentiation repertoire of neural stem cells

Brian W. Booth; David L. Mack; Andreas Androutsellis-Theotokis; Ronald D. G. McKay; Corinne A. Boulanger; Gilbert H. Smith

A fundamental issue in stem cell biology is whether adult somatic stem cells are capable of accessing alternate tissue sites and continue functioning as stem cells in the new microenvironment. To address this issue relative to neurogenic stem cells in the mouse mammary gland microenvironment, we mixed wild-type mammary epithelial cells (MECs) with bona fide neural stem cells (NSCs) isolated from WAP-Cre/Rosa26R mice and inoculated them into cleared fat pads of immunocompromised females. Hosts were bred 6–8 weeks later and examined postinvolution. This allowed for mammary tissue growth, transient activation of the WAP-Cre gene, recombination, and constitutive expression of LacZ. The NSCs and their progeny contributed to mammary epithelial growth during ductal morphogenesis, and the Rosa26-LacZ reporter gene was activated by WAP-Cre expression during pregnancy. Some NSC-derived LacZ+ cells expressed mammary-specific functions, including milk protein synthesis, whereas others adopted myoepithelial cell fates. Thus, NSCs and their progeny enter mammary epithelium–specific niches and adopt the function of similarly endowed mammary cells. This result supports the conclusion that tissue-specific signals emanating from the stroma and from the differentiated somatic cells of the mouse mammary gland can redirect the NSCs to produce cellular progeny committed to MEC fates.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Targeting neural precursors in the adult brain rescues injured dopamine neurons

Andreas Androutsellis-Theotokis; Maria Adele Rueger; Deric M. Park; Haik Mkhikian; Erica Korb; Steve W. Poser; Stuart Walbridge; Jeeva Munasinghe; Alan P. Koretsky; Russel R. Lonser; Ronald D. G. McKay

In Parkinsons disease, multiple cell types in many brain regions are afflicted. As a consequence, a therapeutic strategy that activates a general neuroprotective response may be valuable. We have previously shown that Notch ligands support neural precursor cells in vitro and in vivo. Here we show that neural precursors express the angiopoietin receptor Tie2 and that injections of angiopoietin2 activate precursors in the adult brain. Signaling downstream of Tie2 and the Notch receptor regulate blood vessel formation. In the adult brain, angiopoietin2 and the Notch ligand Dll4 activate neural precursors with opposing effects on the density of blood vessels. A model of Parkinsons disease was used to show that angiopoietin2 and Dll4 rescue injured dopamine neurons with motor behavioral improvement. A combination of growth factors with little impact on the vasculature retains the ability to stimulate neural precursors and protect dopamine neurons. The cellular and pharmacological basis of the neuroprotective effects achieved by these single treatments merits further analysis.


PLOS ONE | 2010

Angiogenic factors stimulate growth of adult neural stem cells.

Andreas Androutsellis-Theotokis; Maria Adele Rueger; Deric M. Park; Justin D. Boyd; Raji Padmanabhan; Loraine Campanati; Craig V. Stewart; Yann LeFranc; Dietmar Plenz; Stuart Walbridge; Russell R. Lonser; Ronald D. G. McKay

Background The ability to grow a uniform cell type from the adult central nervous system (CNS) is valuable for developing cell therapies and new strategies for drug discovery. The adult mammalian brain is a source of neural stem cells (NSC) found in both neurogenic and non-neurogenic zones but difficulties in culturing these hinders their use as research tools [1], [2], [3], [4], [5], [6]. Methodology/Principal Findings Here we show that NSCs can be efficiently grown in adherent cell cultures when angiogenic signals are included in the medium. These signals include both anti-angiogenic factors (the soluble form of the Notch receptor ligand, Dll4) and pro-angiogenic factors (the Tie-2 receptor ligand, Angiopoietin 2). These treatments support the self renewal state of cultured NSCs and expression of the transcription factor Hes3, which also identifies the cancer stem cell population in human tumors. In an organotypic slice model, angiogenic factors maintain vascular structure and increase the density of dopamine neuron processes. Conclusions/Significance We demonstrate new properties of adult NSCs and a method to generate efficient adult NSC cultures from various central nervous system areas. These findings will help establish cellular models relevant to cancer and regeneration.


Brain Research | 1996

The depolarisation-induced release of [125I]BDNF from brain tissue

Andreas Androutsellis-Theotokis; Wendy J McCormack; H. F. Bradford; Gerald M. Stern; F.Bernardo Pliego-Rivero

The pattern of release of radioactive brain-derived neurotrophic factor ([125I]BDNF) from brain tissue was studied. Rat brain slices from cerebral cortex and synaptosomes from cerebral cortex and hippocampus were preloaded with [125I]BDNF. Depolarising stimulation by veratridine (final conc. 50 microM) and high KCl (final conc. 45 mM) caused a short-term, greatly enhanced depolarisation-induced release of [125I]BDNF during superfusion and batch protocol experiments. The results suggested that the evoked release was independent of the presence of extracellular calcium ions, but dependent on intracellular calcium ion stores, since the intracellular calcium ion chelator BAPTA-AM, but not the extracellular chelator EGTA abolished the high-potassium-induced [125I]BDNF release from synaptosomes. The release was blocked by tetrodotoxin (1 microM) when synaptosomes were stimulated by veratridine or potassium chloride. Short time-fraction (30 s) superfusion experiments showed that the [125I]BDNF release from synaptosomes appeared in two temporal phases.


Hepatology | 2014

Dual role of B7 costimulation in obesity-related nonalcoholic steatohepatitis and metabolic dysregulation.

Antonios Chatzigeorgiou; Kyoung-Jin Chung; Ruben Garcia-Martin; Vasileia‐Ismini Alexaki; Anne Klotzsche-von Ameln; Julia Phieler; David Sprott; Waldemar Kanczkowski; Theodora Tzanavari; Mohktar Bdeir; Sibylle Bergmann; Marc Cartellieri; Michael H. Bachmann; Polyxeni Nikolakopoulou; Andreas Androutsellis-Theotokis; Gabriele Siegert; Stefan R. Bornstein; Michael H. Muders; Louis Boon; Katia Karalis; Esther Lutgens; Triantafyllos Chavakis

The low‐grade inflammatory state present in obesity contributes to obesity‐related metabolic dysregulation, including nonalcoholic steatohepatitis (NASH) and insulin resistance. Intercellular interactions between immune cells or between immune cells and hepatic parenchymal cells contribute to the exacerbation of liver inflammation and steatosis in obesity. The costimulatory molecules, B7.1 and B7.2, are important regulators of cell‐cell interactions in several immune processes; however, the role of B7 costimulation in obesity‐related liver inflammation is unknown. Here, diet‐induced obesity (DIO) studies in mice with genetic inactivation of both B7.1 and B7.2 (double knockout; DKO) revealed aggravated obesity‐related metabolic dysregulation, reduced insulin signalling in the liver and adipose tissue (AT), glucose intolerance, and enhanced progression to steatohepatitis resulting from B7.1/B7.2 double deficiency. The metabolic phenotype of B7.1/B7.2 double deficiency upon DIO was accompanied by increased hepatic and AT inflammation, associated with largely reduced numbers of regulatory T cells (Tregs) in these organs. In order to assess the role of B7 costimulation in DIO in a non‐Treg‐lacking environment, we performed antibody (Ab)‐mediated inhibition of B7 molecules in wild‐type mice in DIO. Antibody‐blockade of both B7.1 and B7.2 improved the metabolic phenotype of DIO mice, which was linked to amelioration of hepatic steatosis and reduced inflammation in liver and AT. Conclusion: Our study demonstrates a dual role of B7 costimulation in the course of obesity‐related sequelae, particularly NASH. The genetic inactivation of B7.1/B7.2 deteriorates obesity‐related liver steatosis and metabolic dysregulation, likely a result of the intrinsic absence of Tregs in these mice, rendering DKO mice a novel murine model of NASH. In contrast, inhibition of B7 costimulation under conditions where Tregs are present may provide a novel therapeutic approach for obesity‐related metabolic dysregulation and, especially, NASH. (Hepatology 2014;60:1196–1210)


Cold Spring Harbor Symposia on Quantitative Biology | 2008

Signaling Pathways Controlling Neural Stem Cells Slow Progressive Brain Disease

Andreas Androutsellis-Theotokis; Maria Adele Rueger; H. Mkhikian; E. Korb; Ronald D. G. McKay

The identification and characterization of multipotent neural precursors open the possibility of transplant therapies, but this approach is complicated by the widespread pathology of many degenerative diseases. Activation of endogenous precursors that support regenerative mechanisms is a possible alternative. We have previously shown that Notch ligands promote stem cell survival in vitro. Here, we show that there is an intimate interaction between insulin and Notch receptor signaling. Notch ligands also expand stem cell numbers in vivo with correlated benefits in brain ischemia. We now show that insulin promotes recovery of injured dopamine neurons in the adult brain. This response suggests that activating survival mechanisms in neural stem cells will promote recovery from progressive degenerative disease.


Scientific Reports | 2013

Hes3 regulates cell number in cultures from glioblastoma multiforme with stem cell characteristics.

Deric M. Park; Jinkyu Jung; Jimmy Masjkur; Stylianos Makrogkikas; Doreen Ebermann; Sarama Saha; Roberta Rogliano; Nicoletta Paolillo; Simone Pacioni; Ron McKay; Steve W. Poser; Andreas Androutsellis-Theotokis

Tumors exhibit complex organization and contain a variety of cell populations. The realization that the regenerative properties of a tumor may be largely confined to a cell subpopulation (cancer stem cell) is driving a new era of anti-cancer research. Cancer stem cells from Glioblastoma Multiforme tumors express markers that are also expressed in non-cancerous neural stem cells, including nestin and Sox2. We previously showed that the transcription factor Hes3 is a marker of neural stem cells, and that its expression is inhibited by JAK activity. Here we show that Hes3 is also expressed in cultures from glioblastoma multiforme which express neural stem cell markers, can differentiate into neurons and glia, and can recapitulate the tumor of origin when transplanted into immunocompromised mice. Similar to observations in neural stem cells, JAK inhibits Hes3 expression. Hes3 RNA interference reduces the number of cultured glioblastoma cells suggesting a novel therapeutic strategy.


PLOS ONE | 2010

Cholera toxin regulates a signaling pathway critical for the expansion of neural stem cell cultures from the fetal and adult rodent brains.

Andreas Androutsellis-Theotokis; Stuart Walbridge; Deric M. Park; Russell R. Lonser; Ronald D. G. McKay

Background New mechanisms that regulate neural stem cell (NSC) expansion will contribute to improved assay systems and the emerging regenerative approach that targets endogenous stem cells. Expanding knowledge on the control of stem cell self renewal will also lead to new approaches for targeting the stem cell population of cancers. Methodology/Principal Findings Here we show that Cholera toxin regulates two recently characterized NSC markers, the Tie2 receptor and the transcription factor Hes3, and promotes the expansion of NSCs in culture. Cholera toxin increases immunoreactivity for the Tie2 receptor and rapidly induces the nuclear localization of Hes3. This is followed by powerful cultured NSC expansion and induction of proliferation both in the presence and absence of mitogen. Conclusions/Significance Our data suggest a new cell biological mechanism that regulates the self renewal and differentiation properties of stem cells, providing a new logic to manipulate NSCs in the context of regenerative disease and cancer.

Collaboration


Dive into the Andreas Androutsellis-Theotokis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven W. Poser

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Jimmy Masjkur

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Ronald D. G. McKay

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Triantafyllos Chavakis

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Deric M. Park

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Polyxeni Nikolakopoulou

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steve W. Poser

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge