Network


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

Hotspot


Dive into the research topics where Katelin F. Hansen is active.

Publication


Featured researches published by Katelin F. Hansen.


PLOS ONE | 2010

Transgenic miR132 Alters Neuronal Spine Density and Impairs Novel Object Recognition Memory

Katelin F. Hansen; Kensuke Sakamoto; Gary A. Wayman; Soren Impey; Karl Obrietan

Inducible gene expression plays a central role in neuronal plasticity, learning, and memory, and dysfunction of the underlying molecular events can lead to severe neuronal disorders. In addition to coding transcripts (mRNAs), non-coding microRNAs (miRNAs) appear to play a role in these processes. For instance, the CREB-regulated miRNA miR132 has been shown to affect neuronal structure in an activity-dependent manner, yet the details of its physiological effects and the behavioral consequences in vivo remain unclear. To examine these questions, we employed a transgenic mouse strain that expresses miR132 in forebrain neurons. Morphometric analysis of hippocampal neurons revealed that transgenic miR132 triggers a marked increase in dendritic spine density. Additionally, miR132 transgenic mice exhibited a decrease in the expression of MeCP2, a protein implicated in Rett Syndrome and other disorders of mental retardation. Consistent with these findings, miR132 transgenic mice displayed significant deficits in novel object recognition. Together, these data support a role for miR132 as a regulator of neuronal structure and function, and raise the possibility that dysregulation of miR132 could contribute to an array of cognitive disorders.


Brain Structure & Function | 2013

miRNA-132: a dynamic regulator of cognitive capacity

Katelin F. Hansen; Kate Karelina; Kensuke Sakamoto; Gary A. Wayman; Soren Impey; Karl Obrietan

Within the central nervous system, microRNAs have emerged as important effectors of an array of developmental, physiological, and cognitive processes. Along these lines, the CREB-regulated microRNA miR-132 has been shown to influence neuronal maturation via its effects on dendritic arborization and spinogenesis. In the mature nervous system, dysregulation of miR-132 has been suggested to play a role in a number of neurocognitive disorders characterized by aberrant synaptogenesis. However, little is known about the inducible expression and function of miR-132 under normal physiological conditions in vivo. Here, we begin to explore this question within the context of learning and memory. Using in situ hybridization, we show that the presentation of a spatial memory task induced a significant ~1.5-fold increase in miR-132 expression within the CA1, CA3, and GCL excitatory cell layers of the hippocampus. To examine the role of miR-132 in hippocampal-dependent learning and memory, we employ a doxycycline-regulated miR-132 transgenic mouse strain to drive varying levels of transgenic miR-132 expression. These studies revealed that relatively low levels of transgenic miR-132 expression, paralleling the level of expression in the hippocampus following a spatial memory task, significantly enhanced cognitive capacity. In contrast, higher (supra-physiological) levels of miR-132 (>3-fold) inhibited learning. Interestingly, both the impaired cognition and elevated levels of dendritic spines resulting from supra-physiological levels of transgenic miR-132 were reversed by doxycycline suppression of transgene expression. Together, these data indicate that miR-132 functions as a key activity-dependent regulator of cognition, and that miR-132 expression must be maintained within a limited range to ensure normal learning and memory formation.


Journal of Biological Rhythms | 2010

CREB Influences Timing and Entrainment of the SCN Circadian Clock

Boyoung Lee; Aiqing Li; Katelin F. Hansen; Ruifeng Cao; Jae Hwa Yoon; Karl Obrietan

The transcriptional feedback circuit, which is at the core of the suprachiasmatic nucleus (SCN) circadian (i.e., 24 h) clock, is tightly coupled to both external entrainment cues, such as light, as well as rhythmic cues that arise on a system-wide level within the SCN. One potential signaling pathway by which these cues are conveyed to the molecular clock is the CREB/CRE transcriptional cascade. In this study, we employed a tetracycline-inducible CREB repressor mouse strain, in which approximately 60% of the SCN neurons express the transgene, to test CREB functionality in the clock and its effects on overt rhythmicity. We show that attenuated CREB signaling in the SCN led to a significant reduction in light-evoked clock entrainment. An examination of circadian timing revealed that CREB repressor mice exhibited normal free-running rhythms in the absence of external lighting cues. However, under conditions of constant light, which typically leads to a lengthening of the circadian period, CREB repressor mice exhibited a dramatic arrhythmic phenotype, which could be reversed with doxycycline. At a cellular level, the repression of CREB led to a significant reduction in both the expression of the circadian clock proteins PERIOD1 and PERIOD2 and the clock output hormones AVP and VIP. Together, these data support the idea that the CRE transcriptional pathway orchestrates transcriptional events that are essential for both the maintenance of SCN timing and light entrainment of the circadian clock.


Scientific Reports | 2015

Profiling status epilepticus-induced changes in hippocampal RNA expression using high-throughput RNA sequencing

Katelin F. Hansen; Kensuke Sakamoto; Carl Pelz; Soren Impey; Karl Obrietan

Status epilepticus (SE) is a life-threatening condition that can give rise to a number of neurological disorders, including learning deficits, depression, and epilepsy. Many of the effects of SE appear to be mediated by alterations in gene expression. To gain deeper insight into how SE affects the transcriptome, we employed the pilocarpine SE model in mice and Illumina-based high-throughput sequencing to characterize alterations in gene expression from the induction of SE, to the development of spontaneous seizure activity. While some genes were upregulated over the entire course of the pathological progression, each of the three sequenced time points (12-hour, 10-days and 6-weeks post-SE) had a largely unique transcriptional profile. Hence, genes that regulate synaptic physiology and transcription were most prominently altered at 12-hours post-SE; at 10-days post-SE, marked changes in metabolic and homeostatic gene expression were detected; at 6-weeks, substantial changes in the expression of cell excitability and morphogenesis genes were detected. At the level of cell signaling, KEGG analysis revealed dynamic changes within the MAPK pathways, as well as in CREB-associated gene expression. Notably, the inducible expression of several noncoding transcripts was also detected. These findings offer potential new insights into the cellular events that shape SE-evoked pathology.


PLOS ONE | 2013

A Genome-Wide Screen of CREB Occupancy Identifies the RhoA Inhibitors Par6C and Rnd3 as Regulators of BDNF-Induced Synaptogenesis

Carl Pelz; Hideaki Ando; Mingyan Zhu; Monika A. Davare; Talley J. Lambert; Katelin F. Hansen; Karl Obrietan; Suzanne M. Appleyard; Soren Impey; Gary A. Wayman

Neurotrophin-regulated gene expression is believed to play a key role in long-term changes in synaptic structure and the formation of dendritic spines. Brain-derived neurotrophic factor (BDNF) has been shown to induce increases in dendritic spine formation, and this process is thought to function in part by stimulating CREB-dependent transcriptional changes. To identify CREB-regulated genes linked to BDNF-induced synaptogenesis, we profiled transcriptional occupancy of CREB in hippocampal neurons. Interestingly, de novo motif analysis of hippocampal ChIP-Seq data identified a non-canonical CRE motif (TGGCG) that was enriched at CREB target regions and conferred CREB-responsiveness. Because cytoskeletal remodeling is an essential element of the formation of dendritic spines, within our screens we focused our attention on genes previously identified as inhibitors of RhoA GTPase. Bioinformatic analyses identified dozens of candidate CREB target genes known to regulate synaptic architecture and function. We showed that two of these, the RhoA inhibitors Par6C (Pard6A) and Rnd3 (RhoE), are BDNF-induced CREB-regulated genes. Interestingly, CREB occupied a cluster of non-canonical CRE motifs in the Rnd3 promoter region. Lastly, we show that BDNF-stimulated synaptogenesis requires the expression of Par6C and Rnd3, and that overexpression of either protein is sufficient to increase synaptogenesis. Thus, we propose that BDNF can regulate formation of functional synapses by increasing the expression of the RhoA inhibitors, Par6C and Rnd3. This study shows that genome-wide analyses of CREB target genes can facilitate the discovery of new regulators of synaptogenesis.


Genome Medicine | 2011

MicroRNAs: a potential interface between the circadian clock and human health

Katelin F. Hansen; Kensuke Sakamoto; Karl Obrietan

The biochemical activity of a stunning diversity of cell types and organ systems is shaped by a 24-hour (circadian) clock. This rhythmic drive to a good deal of the transcriptome (up to 15% of all coding genes) imparts circadian modulation over a wide range of physiological and behavioral processes (from cell division to cognition). Further, dysregulation of the clock has been implicated in the pathogenesis of a large and diverse array of disorders, such as hypertension, cancer and depression. Indeed, the possibility of utilizing therapeutic approaches that target clock physiology (that is, chronotherapy) has gained broad interest. However, a deeper understanding of the underlying molecular mechanisms that modulate the clock, and give rise to organ-specific clock transcriptomes, will be required to fully realize the power of chronotherapies. Recently, microRNAs have emerged as significant players in circadian clock timing, thus raising the possibility that clock-controlled microRNAs could contribute to disorders of the human circadian timing system. Here, we highlight recent work revealing a key role for microRNAs in clock physiology, and discuss potential approaches to unlocking their utility as effectors of circadian physiology and pathophysiology.


Neuropsychiatric Disease and Treatment | 2013

MicroRNA as therapeutic targets for treatment of depression

Katelin F. Hansen; Karl Obrietan

Depression is a potentially life-threatening mental disorder affecting approximately 300 million people worldwide. Despite much effort, the molecular underpinnings of clinical depression remain poorly defined, and current treatments carry limited therapeutic efficacy and potentially burdensome side effects. Recently, small noncoding RNA molecules known as microRNA (miRNA) have gained prominence as a target for therapeutic intervention, given their capacity to regulate neuronal physiology. Further, mounting evidence suggests a prominent role for miRNA in depressive molecular signaling. Recent studies have demonstrated that dysregulation of miRNA expression occurs in animal models of depression, and in the post-mortem tissue of clinically depressed patients. Investigations into depression-associated miRNA disruption reveals dramatic effects on downstream targets, many of which are thought to contribute to depressive symptoms. Furthermore, selective serotonin reuptake inhibitors, as well as other antidepressant drugs, have the capacity to reverse aberrant depressive miRNA expression and their downstream targets. Given the powerful effects that miRNA have on the central nervous system transcriptome, and the aforementioned studies, there is a compelling rationale to begin to assess the potential contribution of miRNA to depressive etiology. Here, we review the molecular biology of miRNA, our current understanding of miRNA in relation to clinical depression, and the utility of targeting miRNA for antidepressant treatment.


Learning & Memory | 2016

Targeted deletion of miR-132/-212 impairs memory and alters the hippocampal transcriptome

Katelin F. Hansen; Kensuke Sakamoto; Sydney Aten; Kaitlin H. Snider; Jacob Loeser; Andrea M. Hesse; Chloe E. Page; Carl Pelz; J. Simon C. Arthur; Soren Impey; Karl Obrietan

miR-132 and miR-212 are structurally related microRNAs that have been found to exert powerful modulatory effects within the central nervous system (CNS). Notably, these microRNAs are tandomly processed from the same noncoding transcript, and share a common seed sequence: thus it has been difficult to assess the distinct contribution of each microRNA to gene expression within the CNS. Here, we employed a combination of conditional knockout and transgenic mouse models to examine the contribution of the miR-132/-212 gene locus to learning and memory, and then to assess the distinct effects that each microRNA has on hippocampal gene expression. Using a conditional deletion approach, we show that miR-132/-212 double-knockout mice exhibit significant cognitive deficits in spatial memory, recognition memory, and in tests of novel object recognition. Next, we utilized transgenic miR-132 and miR-212 overexpression mouse lines and the miR-132/-212 double-knockout line to explore the distinct effects of these two miRNAs on the transcriptional profile of the hippocampus. Illumina sequencing revealed that miR-132/-212 deletion increased the expression of 1138 genes; Venn analysis showed that 96 of these genes were also downregulated in mice overexpressing miR-132. Of the 58 genes that were decreased in animals overexpressing miR-212, only four of them were also increased in the knockout line. Functional gene ontology analysis of downregulated genes revealed significant enrichment of genes related to synaptic transmission, neuronal proliferation, and morphogenesis, processes known for their roles in learning, and memory formation. These data, coupled with previous studies, firmly establish a role for the miR-132/-212 gene locus as a key regulator of cognitive capacity. Further, although miR-132 and miR-212 share a seed sequence, these data indicate that these miRNAs do not exhibit strongly overlapping mRNA targeting profiles, thus indicating that these two genes may function in a complex, nonredundant manner to shape the transcriptional profile of the CNS. The dysregulation of miR-132/-212 expression could contribute to signaling mechanisms that are involved in an array of cognitive disorders.


Learning & Memory | 2012

MSK1 regulates environmental enrichment-induced hippocampal plasticity and cognitive enhancement.

Kate Karelina; Katelin F. Hansen; Yun Sik Choi; A. C. DeVries; J. S. C. Arthur; Karl Obrietan

Environmental enrichment (EE) has marked beneficial effects on cognitive capacity. Given the possibility that this form of neuronal plasticity could function via the actuation of the same cellular signaling pathways that underlie learning/memory formation, we examined whether the MAPK cascade effector, mitogen/stress-activated kinase 1 (MSK1), could play a role in this process. MSK1 functions as a key signaling intermediate that couples changes in neuronal activity into inducible gene expression, neuronal plasticity, and learning/memory. Here, we show that MSK1 is expressed in excitatory cell layers of the hippocampus, progenitor cells of the subgranular zone (SGZ), and adult-born immature neurons. MSK1(-/-) mice exhibit reduced spinogenesis and decreased dendritic branching complexity in hippocampal neurons, compared with WT mice. Further, in MSK1(-/-) mice, progenitor cell proliferation within the SGZ was significantly reduced and, correspondingly, the number of immature neurons within the dentate gyrus was significantly reduced. Consistent with prior work, MSK1(-/-) mice displayed deficits in both spatial and recognition memory tasks. Strikingly, cognitive enhancement resulting from a 40-d period of EE was markedly reduced in MSK1(-/-) animals. MSK1(-/-) mice exhibited reduced levels of EE-induced spinogenesis and SGZ progenitor proliferation. Taken together, these data reveal that MSK1 serves as a critical regulator of hippocampal physiology and function and that MSK1 serves as a key conduit by which enriching stimuli augment cellular plasticity and cognition.


Frontiers in Cellular Neuroscience | 2013

MicroRNAs: fundamental regulators of gene expression in major affective disorders and suicidal behavior?

Gianluca Serafini; Maurizio Pompili; Katelin F. Hansen; Karl Obrietan; Yogesh Dwivedi; Mario Amore; Noam Shomron; Paolo Girardi

Major affective disorders are one of the foremost causes of morbidity worldwide; such disabling conditions are also frequently associated with suicidal behavior (Innamorati et al., 2011; Gonda et al., 2012; Serafini et al., 2012). Although many psychopharmacological agents are currently available, in particular for the treatment of major depressive disorder (MDD) (Serafini et al., 2013), our knowledge concerning the molecular and cellular mechanisms underlying this complex condition is still limited. Indeed, even minor alterations in the expression of genes regulating neural and structural plasticity may be crucial to understanding the pathogenesis of major affective disorders (Dwivedi et al., 2009a,b; Serafini et al., 2011, 2012). MiRNAs are gene expression regulators critically affecting brain development that have been investigated as potential biomarkers for the diagnosis, management, treatment, and progression of neuropsychiatric disorders (Machado-Vieira et al., 2010; Saugstad, 2010; Dwivedi, 2011). Several facets of miRNA expression alterations are currently under investigation to gain insight into the pathology of neuronal disorders (Hansen et al., 2007; Lopez et al., 2013): miRNA expression alterations in pathophysiological models of disease (Ziu et al., 2011; Brandenburger et al., 2012); miRNA expression alterations in the blood of patients, most of which represent further downstream or compensatory effects (Schipper et al., 2007; Gallego et al., 2012); and miRNAs and their effectors acting as targets for the action of psychoactive drugs such as antidepressants and mood stabilizers (Zhou et al., 2009; Baudry et al., 2010; Oved et al., 2012). Several miRNAs have emerged as potential mediators of depressive pathophysiology. The existence of polymorphisms in pre-miR-30e (Xu et al., 2010) and pre-miR-182 (Saus et al., 2010) has been associated with an increased risk of major depression. Depressive behavioral responses have been induced by miR-16 up-regulation in the raphe nuclei and hippocampus, with the latter associated with subsequent down-regulation of BNDF (Bai et al., 2012). MiR-16 down-regulation within the locus coeruleus was also induced in depressive mouse models (Launay et al., 2011). Smalheiser et al. (2012) investigated the expression of miRNAs in the prefrontal cortex (specifically in Brodmann Area 9) of 18 antidepressant-free depressed suicide victims, and 17 well-matched non-psychiatric controls, whose information was collected using the psychological autopsy method. In this study, global miRNA expression was significantly down-regulated by 17%, and 24 miRNAs were down-regulated by at least 30%. The authors also found significant down-regulation in an extensive inter-connected network of 21 miRNAs involved in cellular growth and differentiation. Notably, both the global decrease of miRNA expression, as well as its decreased variability, are consistent with hypo-activation of the frontal cortex in depressed subjects. Interestingly, the noted miRNAs have been suggested to be down-regulated in the frontal cortex of rats treated with corticosterone, and therefore, might be crucial in regulating stress-mediated miRNA expression in depressed subjects (Dwivedi et al., unpublished data). Hence, alterations in miRNA expression may be a fundamental event underlying gene network reorganization associated with major depression. Nevertheless, a comprehensive understanding of miRNA networks dysregulated in major depression and induced by antidepressant medications as a function of brain region is currently unknown (Mouillet-Richard et al., 2012). It is also generally poorly understood how miRNA regulation affects cellular signaling networks in these biological processes. Here we provide an overview and critical review of the published work, particularly examining the role of miR-185 in major depression and suicidal behavior.

Collaboration


Dive into the Katelin F. Hansen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary A. Wayman

Washington State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yun Sik Choi

Catholic University of Daegu

View shared research outputs
Researchain Logo
Decentralizing Knowledge