Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
FASEB J ; 33(8): 9235-9249, 2019 08.
Article in English | MEDLINE | ID: mdl-31145643

ABSTRACT

Cancer cells can switch between signaling pathways to regulate growth under different conditions. In the tumor microenvironment, this likely helps them evade therapies that target specific pathways. We must identify all possible states and utilize them in drug screening programs. One such state is characterized by expression of the transcription factor Hairy and Enhancer of Split 3 (HES3) and sensitivity to HES3 knockdown, and it can be modeled in vitro. Here, we cultured 3 primary human brain cancer cell lines under 3 different culture conditions that maintain low, medium, and high HES3 expression and characterized gene regulation and mechanical phenotype in these states. We assessed gene expression regulation following HES3 knockdown in the HES3-high conditions. We then employed a commonly used human brain tumor cell line to screen Food and Drug Administration (FDA)-approved compounds that specifically target the HES3-high state. We report that cells from multiple patients behave similarly when placed under distinct culture conditions. We identified 37 FDA-approved compounds that specifically kill cancer cells in the high-HES3-expression conditions. Our work reveals a novel signaling state in cancer, biomarkers, a strategy to identify treatments against it, and a set of putative drugs for potential repurposing.-Poser, S. W., Otto, O., Arps-Forker, C., Ge, Y., Herbig, M., Andree, C., Gruetzmann, K., Adasme, M. F., Stodolak, S., Nikolakopoulou, P., Park, D. M., Mcintyre, A., Lesche, M., Dahl, A., Lennig, P., Bornstein, S. R., Schroeck, E., Klink, B., Leker, R. R., Bickle, M., Chrousos, G. P., Schroeder, M., Cannistraci, C. V., Guck, J., Androutsellis-Theotokis, A. Controlling distinct signaling states in cultured cancer cells provides a new platform for drug discovery.


Subject(s)
Glioblastoma/metabolism , Repressor Proteins/metabolism , Cell Line, Tumor , Drug Discovery , Gene Expression Profiling , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Glioblastoma/genetics , Humans , RNA Interference , Repressor Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology
2.
J Biol Chem ; 289(51): 35503-16, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25371201

ABSTRACT

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.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/genetics , Gene Expression , Insulin/metabolism , Islets of Langerhans/metabolism , Transcription Factors/genetics , Adult , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Exenatide , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/genetics , Insulin Secretion , Insulinoma/genetics , Insulinoma/metabolism , Insulinoma/pathology , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Obese , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides/pharmacology , RNA Interference , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Venoms/pharmacology
3.
Sci Rep ; 8(1): 11335, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054579

ABSTRACT

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 ß-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.


Subject(s)
Aging/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Diet, High-Fat , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Metformin/administration & dosage , Nerve Tissue Proteins/metabolism , Streptozocin/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/drug effects , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phenotype , Repressor Proteins
4.
Sci Rep ; 7: 43946, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287094

ABSTRACT

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.

5.
Diabetes ; 65(2): 314-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26798118

ABSTRACT

Loss of insulin-producing pancreatic islet ß-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.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Neural Stem Cells/physiology , Pancreas/physiology , Regeneration/physiology , Animals , Cell Differentiation/physiology , Hedgehog Proteins/physiology , Humans , Mice , Mice, Nude , Organogenesis/physiology , Pancreas/embryology , Signal Transduction/physiology
6.
Brain Res ; 1642: 124-130, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27018293

ABSTRACT

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 Parkinson's 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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Demyelinating Diseases/genetics , Gene Expression Regulation , Motor Cortex/metabolism , Myelin Sheath/genetics , Nerve Tissue Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Culture Techniques , Culture Media, Conditioned , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , Repressor Proteins
7.
Stem Cells Transl Med ; 4(11): 1251-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26371344

ABSTRACT

UNLABELLED: 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 ß-like cells). Specifically for acinar-to-ß-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. SIGNIFICANCE: Reprogramming is a powerful approach to change cell identity, with implications in both basic and applied biology. Most efforts involve the forced expression of key transcription factors, but recently, success has been reported with manipulating signal transduction pathways that might intercept them. It is important to start connecting the function of the classic reprogramming genes to signaling pathways that also mediate reprogramming, unifying the sciences of signal transduction, stem cell biology, and epigenetics. Neural stem cell studies have revealed the operation of noncanonical signaling pathways that are now appreciated to also operate during reprogramming, offering new mechanistic explanations.


Subject(s)
Cellular Reprogramming , DNA-Binding Proteins/biosynthesis , Neural Stem Cells/metabolism , STAT3 Transcription Factor/biosynthesis , Signal Transduction , Transcription Factors/biosynthesis , Animals , DNA-Binding Proteins/genetics , Humans , Neural Stem Cells/cytology , Repressor Proteins , STAT3 Transcription Factor/genetics , Transcription Factors/genetics
8.
Stem Cell Res Ther ; 5(6): 127, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25688994

ABSTRACT

There is a growing interest in the therapeutic utility of compounds derived from Curcuma longa, an herb of the Zingiberaceae family that has been part of traditional medicine for centuries. Recent reports suggest that bioactive compounds isolated from the rhizome of these plants can address two key aspects of brain injury following stroke that must be dealt with for functional recovery to occur: the moderation of neuroinflammation, and the mobilization of endogenous stem cells resident in the nervous system. Defining their mechanism of action remains a question, but emerging evidence may point towards one shared with more classic modulators of neural stem cell proliferation and survival.


Subject(s)
Cell Proliferation/drug effects , Ketones/administration & dosage , Neural Stem Cells/physiology , Neuroprotective Agents/administration & dosage , Sesquiterpenes/administration & dosage , Animals , Male
9.
Methods Mol Biol ; 1213: 293-302, 2014.
Article in English | MEDLINE | ID: mdl-25173392

ABSTRACT

In order to establish novel therapeutic paradigms and advance the field of regenerative medicine, methods for their effective implementation as well as rigorous assessment of outcomes are critical. This is especially evident and challenging in the context of treating complex and devastating neurodegenerative disorders, such as Parkinson's disease, multiple sclerosis, and ischemic stroke. Stem cell-based approaches offer great promise in addressing these conditions. Here, we demonstrate an approach for identifying factors that mobilize endogenous neural stem cells in the repair and recovery of the central nervous system of rodents, involving site-specific administration of growth factors that activate particular signal transduction pathways, and that allows for the assessment of outcome utilizing magnetic resonance imaging and immunohistochemistry.


Subject(s)
Nerve Regeneration , Neural Stem Cells/cytology , Regenerative Medicine , Stem Cell Transplantation , Animals , Brain/metabolism , Brain/pathology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/pathology , Immunohistochemistry , Magnetic Resonance Imaging , Male , Neural Stem Cells/metabolism , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/therapy , Rats , Regenerative Medicine/methods
10.
Front Biosci (Landmark Ed) ; 19(4): 718-26, 2014 01 01.
Article in English | MEDLINE | ID: mdl-24389215

ABSTRACT

Disrupting the regenerative capacity of tumorigenic cells is a major focus in medicine. These regenerative properties are carried by a subpopulation of cells within the tumor, termed cancer stem cells. Current therapies don't effectively tackle the disease suggesting these cells employ yet unidentified molecular mechanisms allowing them to evade targeting. Recent observations in neural stem cells reveal an extraordinary plasticity in the signaling pathways they utilize to grow. These findings are being extended to the cancer stem cell field, illuminating conceptually novel treatment strategies. Tumorigenic cells can make use of distinct, even opposing pathways, including JAK/STAT and the non-canonical STAT3-Ser/Hes3 signaling axis. This plasticity may not be confined to the cancer stem cell population, but may be shared by various cell types within the tumor, blurring the line distinguishing cancer stem cells from other tumor cell types. The implications to anti-cancer medicine are highly significant, since these findings demonstrate that inhibiting one cell growth pathway may actually enhance the activity of alternative ones. Drug discovery programs will also benefit from these concepts.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Serine/metabolism , Signal Transduction , Transcription Factors/metabolism , Humans , Repressor Proteins
11.
J Vis Exp ; (81): e50880, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24300750

ABSTRACT

Recent work demonstrates that central nervous system (CNS) regeneration and tumorigenesis involves populations of stem cells (SCs) resident within the adult brain. However, the mechanisms these normally quiescent cells employ to ensure proper functioning of neural networks, as well as their role in recovery from injury and mitigation of neurodegenerative processes are little understood. These cells reside in regions referred to as "niches" that provide a sustaining environment involving modulatory signals from both the vascular and immune systems. The isolation, maintenance, and differentiation of CNS SCs under defined culture conditions which exclude unknown factors, makes them accessible to treatment by pharmacological or genetic means, thus providing insight into their in vivo behavior. Here we offer detailed information on the methods for generating cultures of CNS SCs from distinct regions of the adult brain and approaches to assess their differentiation potential into neurons, astrocytes, and oligodendrocytes in vitro. This technique yields a homogeneous cell population as a monolayer culture that can be visualized to study individual SCs and their progeny. Furthermore, it can be applied across different animal model systems and clinical samples, being used previously to predict regenerative responses in the damaged adult nervous system.


Subject(s)
Brain/cytology , Cytological Techniques/methods , Neural Stem Cells/cytology , Animals , Rats
12.
Front Physiol ; 4: 273, 2013.
Article in English | MEDLINE | ID: mdl-24101906

ABSTRACT

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.

SELECTION OF CITATIONS
SEARCH DETAIL