Network


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

Hotspot


Dive into the research topics where Steven W. Poser is active.

Publication


Featured researches published by Steven W. Poser.


Journal of Biological Chemistry | 1998

STIMULATION OF TYPE 1 AND TYPE 8 CA2+/CALMODULIN-SENSITIVE ADENYLYL CYCLASES BY THE GS-COUPLED 5-HYDROXYTRYPTAMINE SUBTYPE 5-HT7A RECEPTOR

Lauren P. Baker; Mark D. Nielsen; Soren Impey; Mark A. Metcalf; Steven W. Poser; Guy C.-K. Chan; Karl Obrietan; Mark W. Hamblin; Daniel R. Storm

The neurotransmitter serotonin (5-hydroxytryptamine, 5-HT) plays an important regulatory role in developing and adult nervous systems. With the exception of the 5-HT3 receptor, all of the cloned serotonin receptors belong to the G protein-coupled receptor superfamily. Subtypes 5-HT6 and 5-HT7 couple to stimulation of adenylyl cyclases through Gs and display high affinities for antipsychotic and antidepressant drugs. In the brain, mRNA for 5-HT6 is found at high levels in the hippocampus, striatum, and nucleus accumbens. 5-HT7 mRNA is most abundant in the hippocampus, neocortex, and hypothalamus. To better understand how serotonin might control cAMP levels in the brain, we coexpressed 5-HT6 or 5-HT7A receptors with specific isoforms of adenylyl cyclase in HEK 293 cells. The 5-HT6receptor functioned as a typical Gs-coupled receptor in that it stimulated AC5, a Gs-sensitive adenylyl cyclase, but not AC1 or AC8, calmodulin (CaM)-stimulated adenylyl cyclases that are not activated by Gs-coupled receptors in vivo. Surprisingly, serotonin activation of 5-HT7Astimulated AC1 and AC8 by increasing intracellular Ca2+. 5-HT also increased intracellular Ca2+ in primary neuron cultures. These data define a novel mechanism for the regulation of intracellular cAMP by serotonin.


Journal of Biological Chemistry | 1996

Differential Regulation of Type I and Type VIII Ca2+-stimulated Adenylyl Cyclases by Gi-coupled Receptors in Vivo

Mark D. Nielsen; Guy C.-K. Chan; Steven W. Poser; Daniel R. Storm

Coupling of intracellular Ca2+ to cAMP increases may be important for some forms of synaptic plasticity. The type I adenylyl cyclase (I-AC) is a neural-specific, Ca2+-stimulated enzyme that couples intracellular Ca2+ to cAMP increases. Since optimal cAMP levels may be crucial for some types of synaptic plasticity, mechanisms for inhibition of Ca2+-stimulated adenylyl cyclases may also be important for neuroplasticity. Here we report that Ca2+ stimulation of I-AC is inhibited by activation of Gi-coupled somatostatin and dopamine D2L receptors. This inhibition is due primarily to Giα and not βγ subunits since coexpression of βγ-binding proteins with I-AC did not affect somatostatin inhibition. However, βγ released from Gs did inhibit I-AC, indicating that the enzyme can be inhibited by βγ in vivo. Interestingly, type VIII adenylyl cyclase (VIII-AC), another Ca2+-stimulated adenylyl cyclase, was not inhibited by Gi-coupled receptors. These data indicate that I-AC and VIII-AC are differentially regulated by Gi-coupled receptors and provide distinct mechanisms for interactions between the Ca2+ and cAMP signal transduction systems. We propose that I-AC may be particularly important for synaptic plasticity that depends upon rapid and transient cAMP increases, whereas VIII-AC may contribute to transcriptional-dependent synaptic plasticity that is dependent upon prolonged, Ca2+-stimulated cAMP increases.


Journal of Biological Chemistry | 2014

Hes3 is expressed in the adult pancreatic islet and regulates gene expression, cell growth, and insulin release

Jimmy Masjkur; Carina Arps-Forker; Steven W. Poser; Polyxeni Nikolakopoulou; Louiza Toutouna; Ramu Chenna; Triantafyllos Chavakis; Antonis Chatzigeorgiou; Lan-Sun Chen; Anna Dubrovska; Pratik Choudhary; Ingo Uphues; Michael Mark; Stefan R. Bornstein; Andreas Androutsellis-Theotokis

Background: The transcription factor Hes3 regulates the growth of neural and brain cancer stem cells. Results: Hes3 regulates growth, gene expression, evoked insulin release in cultured insulinoma cells, and sensitivity to streptozotocin in vivo. Conclusion: Hes3 is a novel regulator of cellular functions of importance in diabetes. Significance: Introducing Hes3 and its regulators in diabetes research may provide new opportunities for the design of novel therapeutics. The transcription factor Hes3 is a component of a signaling pathway that supports the growth of neural stem cells with profound consequences in neurodegenerative disease models. Here we explored whether Hes3 also regulates pancreatic islet cells. We showed that Hes3 is expressed in human and rodent pancreatic islets. In mouse islets it co-localizes with alpha and beta cell markers. We employed the mouse insulinoma cell line MIN6 to perform in vitro characterization and functional studies in conditions known to modulate Hes3 based upon our previous work using neural stem cell cultures. In these conditions, cells showed elevated Hes3 expression and nuclear localization, grew efficiently, and showed higher evoked insulin release responses, compared with serum-containing conditions. They also exhibited higher expression of the transcription factor Pdx1 and insulin. Furthermore, they were responsive to pharmacological treatments with the GLP-1 analog Exendin-4, which increased nuclear Hes3 localization. We employed a transfection approach to address specific functions of Hes3. Hes3 RNA interference opposed cell growth and affected gene expression as revealed by DNA microarrays. Western blotting and PCR approaches specifically showed that Hes3 RNA interference opposes the expression of Pdx1 and insulin. Hes3 overexpression (using a Hes3-GFP fusion construct) confirmed a role of Hes3 in regulating Pdx1 expression. Hes3 RNA interference reduced evoked insulin release. Mice lacking Hes3 exhibited increased islet damage by streptozotocin. These data suggest roles of Hes3 in pancreatic islet function.


Scientific Reports | 2017

Enlightening discriminative network functional modules behind Principal Component Analysis separation in differential-omic science studies

Sara Ciucci; Yan Ge; Claudio Durán; Alessandra Palladini; Víctor Jiménez-Jiménez; Luisa María Martínez-Sánchez; Yuting Wang; Susanne Sales; Andrej Shevchenko; Steven W. Poser; Maik Herbig; Oliver Otto; Andreas Androutsellis-Theotokis; Jochen Guck; Mathias J. Gerl; Carlo Vittorio Cannistraci

Omic science is rapidly growing and one of the most employed techniques to explore differential patterns in omic datasets is principal component analysis (PCA). However, a method to enlighten the network of omic features that mostly contribute to the sample separation obtained by PCA is missing. An alternative is to build correlation networks between univariately-selected significant omic features, but this neglects the multivariate unsupervised feature compression responsible for the PCA sample segregation. Biologists and medical researchers often prefer effective methods that offer an immediate interpretation to complicated algorithms that in principle promise an improvement but in practice are difficult to be applied and interpreted. Here we present PC-corr: a simple algorithm that associates to any PCA segregation a discriminative network of features. Such network can be inspected in search of functional modules useful in the definition of combinatorial and multiscale biomarkers from multifaceted omic data in systems and precision biomedicine. We offer proofs of PC-corr efficacy on lipidomic, metagenomic, developmental genomic, population genetic, cancer promoteromic and cancer stem-cell mechanomic data. Finally, PC-corr is a general functional network inference approach that can be easily adopted for big data exploration in computer science and analysis of complex systems in physics.


Frontiers in Physiology | 2013

The STAT3-Ser/Hes3 signaling axis: an emerging regulator of endogenous regeneration and cancer growth

Steven W. Poser; Deric M. Park; Andreas Androutsellis-Theotokis

Stem cells, by definition, are able to both self-renew (give rise to more cells of their own kind) and demonstrate multipotential (the ability to differentiate into multiple cell types). To accommodate this unique dual ability, stem cells interpret signal transduction pathways in specialized ways. Notable examples include canonical and non-canonical branches of the Notch signaling pathway, with each controlling different downstream targets (e.g., Hes1 vs. Hes3) and promoting either differentiation or self-renewal. Similarly, stem cells utilize STAT3 signaling uniquely. Most mature cells studied thus far rely on tyrosine phosphorylation (STAT3-Tyr) to promote survival and growth; in contrast, STAT3-Tyr induces the differentiation of neural stem cells (NSCs). NSCs use an alternative phosphorylation site, STAT3-Ser, to regulate survival and growth, a site that is largely redundant for this function in most other cell types. STAT3-Ser regulates Hes3, and together they form a convergence point for several signals, including Notch, Tie2, and insulin receptor activation. Disregulation and manipulation of the STAT3-Ser/Hes3 signaling pathway is important in both tumorigenesis and regenerative medicine, and worthy of extensive study.


Diabetes | 2016

Endocrine Pancreas Development and Regeneration: Noncanonical Ideas From Neural Stem Cell Biology

Jimmy Masjkur; Steven W. Poser; Polyxeni Nikolakopoulou; George P. Chrousos; Ronald D. G. McKay; Stefan R. Bornstein; Peter M. Jones; Andreas Androutsellis-Theotokis

Loss of insulin-producing pancreatic islet β-cells is a hallmark of type 1 diabetes. Several experimental paradigms demonstrate that these cells can, in principle, be regenerated from multiple endogenous sources using signaling pathways that are also used during pancreas development. A thorough understanding of these pathways will provide improved opportunities for therapeutic intervention. It is now appreciated that signaling pathways should not be seen as “on” or “off” but that the degree of activity may result in wildly different cellular outcomes. In addition to the degree of operation of a signaling pathway, noncanonical branches also play important roles. Thus, a pathway, once considered as “off” or “low” may actually be highly operational but may be using noncanonical branches. Such branches are only now revealing themselves as new tools to assay them are being generated. A formidable source of noncanonical signal transduction concepts is neural stem cells because these cells appear to have acquired unusual signaling interpretations to allow them to maintain their unique dual properties (self-renewal and multipotency). We discuss how such findings from the neural field can provide a blueprint for the identification of new molecular mechanisms regulating pancreatic biology, with a focus on Notch, Hes/Hey, and hedgehog pathways.


Stem Cells Translational Medicine | 2015

Concise Review: Reprogramming, Behind the Scenes: Noncanonical Neural Stem Cell Signaling Pathways Reveal New, Unseen Regulators of Tissue Plasticity With Therapeutic Implications

Steven W. Poser; Josh G. Chenoweth; Carlo Colantuoni; Jimmy Masjkur; George P. Chrousos; Stefan R. Bornstein; Ronald D. G. McKay; Andreas Androutsellis-Theotokis

Interest is great in the new molecular concepts that explain, at the level of signal transduction, the process of reprogramming. Usually, transcription factors with developmental importance are used, but these approaches give limited information on the signaling networks involved, which could reveal new therapeutic opportunities. Recent findings involving reprogramming by genetic means and soluble factors with well‐studied downstream signaling mechanisms, including signal transducer and activator of transcription 3 (STAT3) and hairy and enhancer of split 3 (Hes3), shed new light into the molecular mechanisms that might be involved. We examine the appropriateness of common culture systems and their ability to reveal unusual (noncanonical) signal transduction pathways that actually operate in vivo. We then discuss such novel pathways and their importance in various plastic cell types, culminating in their emerging roles in reprogramming mechanisms. We also discuss a number of reprogramming paradigms (mouse induced pluripotent stem cells, direct conversion to neural stem cells, and in vivo conversion of acinar cells to β‐like cells). Specifically for acinar‐to‐β‐cell reprogramming paradigms, we discuss the common view of the underlying mechanism (involving the Janus kinase‐STAT pathway that leads to STAT3‐tyrosine phosphorylation) and present alternative interpretations that implicate STAT3‐serine phosphorylation alone or serine and tyrosine phosphorylation occurring in sequential order. The implications for drug design and therapy are important given that different phosphorylation sites on STAT3 intercept different signaling pathways. We introduce a new molecular perspective in the field of reprogramming with broad implications in basic, biotechnological, and translational research.


Brain Research | 2016

Hes3 expression in the adult mouse brain is regulated during demyelination and remyelination

Louiza Toutouna; Polyxeni Nikolakopoulou; Steven W. Poser; Jimmy Masjkur; Carina Arps-Forker; Maria Troullinaki; Sylvia Grossklaus; Viktoria Bosak; Ulrike Friedrich; Tjalf Ziemssen; Stefan R. Bornstein; Triantafyllos Chavakis; Andreas Androutsellis-Theotokis

Hes3 is a component of the STAT3-Ser/Hes3 Signaling Axis controlling the growth and survival of neural stem cells and other plastic cells. Pharmacological activation of this pathway promotes neuronal rescue and behavioral recovery in models of ischemic stroke and Parkinsons disease. Here we provide initial observations implicating Hes3 in the cuprizone model of demyelination and remyelination. We focus on the subpial motor cortex of mice because we detected high Hes3 expression. This area is of interest as it is impacted both in human demyelinating diseases and in the cuprizone model. We report that Hes3 expression is reduced at peak demyelination and is partially restored within 1 week after cuprizone withdrawal. This raises the possibility of Hes3 involvement in demyelination/remyelination that may warrant additional research. Supporting a possible role of Hes3 in the maintenance of oligodendrocyte markers, a Hes3 null mouse strain shows lower levels of myelin basic protein in undamaged adult mice, compared to wild-type controls. We also present a novel method for culturing the established oligodendrocyte progenitor cell line oli-neu in a manner that maintains Hes3 expression as well as its self-renewal and differentiation potential, offering an experimental tool to study Hes3. Based upon this approach, we identify a Janus kinase inhibitor and dbcAMP as powerful inducers of Hes3 gene expression. We provide a new biomarker and cell culture method that may be of interest in demyelination/remyelination research.


Hormone and Metabolic Research | 2016

STAT3-Ser/Hes3 Signaling: A New Molecular Component of the Neuroendocrine System?

Polyxeni Nikolakopoulou; Steven W. Poser; Jimmy Masjkur; M Fernandez Rubin de Celis; Louiza Toutouna; Cynthia L. Andoniadou; Ronald D. G. McKay; George P. Chrousos; Monika Ehrhart-Bornstein; Stefan R. Bornstein; Andreas Androutsellis-Theotokis

The endocrine system involves communication among different tissues in distinct organs, including the pancreas and components of the Hypothalamic-Pituitary-Adrenal Axis. The molecular mechanisms underlying these complex interactions are a subject of intense study as they may hold clues for the progression and treatment of a variety of metabolic and degenerative diseases. A plethora of signaling pathways, activated by hormones and other endocrine factors have been implicated in this communication. Recent advances in the stem cell field introduce a new level of complexity: adult progenitor cells appear to utilize distinct signaling pathways than the more mature cells in the tissue they co-reside. It is therefore important to elucidate the signal transduction requirements of adult progenitor cells in addition to those of mature cells. Recent evidence suggests that a common non-canonical signaling pathway regulates adult progenitors in several different tissues, rendering it as a potentially valuable starting point to explore their biology. The STAT3-Ser/Hes3 Signaling Axis was first identified as a major regulator of neural stem cells and, subsequently, cancer stem cells. In the endocrine/neuroendocrine system, this pathway operates on several levels, regulating other types of plastic cells: (a) it regulates pancreatic islet cell function and insulin release; (b) insulin in turn activates the pathway in broadly distributed neural progenitors and possibly also hypothalamic tanycytes, cells with important roles in the control of the adrenal gland; (c) adrenal progenitors themselves operate this pathway. The STAT3-Ser/Hes3 Signaling Axis therefore deserves additional research in the context of endocrinology.


Scientific Reports | 2018

Streptozotocin-induced β-cell damage, high fat diet, and metformin administration regulate Hes3 expression in the adult mouse brain

Polyxeni Nikolakopoulou; Antonios Chatzigeorgiou; Ioannis Kourtzelis; Louiza Toutouna; Jimmy Masjkur; Carina Arps-Forker; Steven W. Poser; Jan Rozman; Birgit Rathkolb; Juan Antonio Aguilar-Pimentel; Eckhard Wolf; Martin Klingenspor; Markus Ollert; Carsten B. Schmidt-Weber; Helmut Fuchs; Valérie Gailus-Durner; Martin Hrabé de Angelis; Vasiliki Tsata; Laura Sebastian Monasor; Maria Troullinaki; Anke Witt; Vivian Anastasiou; George P. Chrousos; Chun-Xia Yi; Cristina García-Cáceres; Matthias H. Tschöp; Stefan R. Bornstein; Andreas Androutsellis-Theotokis

Diabetes mellitus is a group of disorders characterized by prolonged high levels of circulating blood glucose. Type 1 diabetes is caused by decreased insulin production in the pancreas whereas type 2 diabetes may develop due to obesity and lack of exercise; it begins with insulin resistance whereby cells fail to respond properly to insulin and it may also progress to decreased insulin levels. The brain is an important target for insulin, and there is great interest in understanding how diabetes affects the brain. In addition to the direct effects of insulin on the brain, diabetes may also impact the brain through modulation of the inflammatory system. Here we investigate how perturbation of circulating insulin levels affects the expression of Hes3, a transcription factor expressed in neural stem and progenitor cells that is involved in tissue regeneration. Our data show that streptozotocin-induced β-cell damage, high fat diet, as well as metformin, a common type 2 diabetes medication, regulate Hes3 levels in the brain. This work suggests that Hes3 is a valuable biomarker helping to monitor the state of endogenous neural stem and progenitor cells in the context of diabetes mellitus.

Collaboration


Dive into the Steven W. Poser's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jimmy Masjkur

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Polyxeni Nikolakopoulou

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Louiza Toutouna

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

George P. Chrousos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Carina Arps-Forker

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Soren Impey

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge