Network


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

Hotspot


Dive into the research topics where Jennifer W. Mitchell is active.

Publication


Featured researches published by Jennifer W. Mitchell.


Cell and Tissue Research | 2002

Signaling in the suprachiasmatic nucleus: selectively responsive and integrative

Martha U. Gillette; Jennifer W. Mitchell

Abstract. The suprachiasmatic nucleus (SCN) contains a biological clock that generates timing signals that drive daily rhythms in behaviors and homeostatic functions. In addition to this pacemaker function, the SCN gates its own sensitivity to incoming signals, which permits appropriate temporal adjustment to achieve synchrony with environmental and organismic states. A series of time-domains, in which the SCN restricts its own sensitivity to a limited set of stimuli that adjust clock phase, can be distinguished. Pituitary adenylyl cyclase-activating peptide (PACAP) and cAMP directly reset clock phase during the daytime domain; both cause phase advances only during the clocks day-time domain, but are without effect at night. In contrast, acetylcholine and cGMP analogs phase advance the clock only when applied during the night. Sensitivity to light and glutamate arises concomitant with sensitivity to acetylcholine and cGMP. Light and glutamate cause phase delays in the early night, by elevating intracellular Ca2+ via neuronal ryanodine receptors. In late night, light and glutamate utilize a cGMP-mediated mechanism to induce phase advances. Nocturnal responses of SCN primed by light or glutamate can be modulated by effectors of phase-resetting in daytime, namely, PACAP and cAMP. Finally, the dusk and dawn domains are characterized by sensitivity to the pineal hormone, melatonin, acting through protein kinase C. These changing patterns of sensitivities demonstrate that the circadian clock controls multiple intracellular gates, which ensures that they can be opened selectively only at specific points in the circadian cycle. Discerning the molecular bases of these changes is fundamental to understanding integrative and regulatory mechanisms in the circadian system.


Neuron | 2004

Protein Kinase G Type II Is Required for Night-to-Day Progression of the Mammalian Circadian Clock

Shelley A. Tischkau; Jennifer W. Mitchell; Laura A. Pace; Jessica W. Barnes; Jeffrey A. Barnes; Martha U. Gillette

Circadian clocks comprise a cyclic series of dynamic cellular states, characterized by the changing availability of substrates that alter clock time when activated. To determine whether circadian clocks, like the cell cycle, exhibit regulation by key phosphorylation events, we examined endogenous kinase regulation of timekeeping in the mammalian suprachiasmatic nucleus (SCN). Short-term inhibition of PKG-II but not PKG-Ibeta using antisense oligodeoxynucleotides delayed rhythms of electrical activity and Bmal1 mRNA. Phase resetting was rapid and dynamic; inhibition of PKG-II forced repetition of the last 3.5 hr of the cycle. Chronic inhibition of PKG-II disrupted electrical activity rhythms and tonically increased Bmal1 mRNA. PKG-II-like immunoreactivity was detected after coimmunoprecipitation with CLOCK, and CLOCK was phosphorylated in the presence of active PKG-II. PKG-II activation may define a critical control point for temporal progression into the daytime domain by acting on the positive arm of the transcriptional/translational feedback loop.


PLOS ONE | 2010

Circadian Integration of Glutamatergic Signals by Little SAAS in Novel Suprachiasmatic Circuits

Norman Atkins; Jennifer W. Mitchell; Elena V. Romanova; Daniel J. Morgan; Tara P. Cominski; Jennifer L. Ecker; John E. Pintar; Jonathan V. Sweedler; Martha U. Gillette

Background Neuropeptides are critical integrative elements within the central circadian clock in the suprachiasmatic nucleus (SCN), where they mediate both cell-to-cell synchronization and phase adjustments that cause light entrainment. Forward peptidomics identified little SAAS, derived from the proSAAS prohormone, among novel SCN peptides, but its role in the SCN is poorly understood. Methodology/Principal Findings Little SAAS localization and co-expression with established SCN neuropeptides were evaluated by immunohistochemistry using highly specific antisera and stereological analysis. Functional context was assessed relative to c-FOS induction in light-stimulated animals and on neuronal circadian rhythms in glutamate-stimulated brain slices. We found that little SAAS-expressing neurons comprise the third most abundant neuropeptidergic class (16.4%) with unusual functional circuit contexts. Little SAAS is localized within the densely retinorecipient central SCN of both rat and mouse, but not the retinohypothalamic tract (RHT). Some little SAAS colocalizes with vasoactive intestinal polypeptide (VIP) or gastrin-releasing peptide (GRP), known mediators of light signals, but not arginine vasopressin (AVP). Nearly 50% of little SAAS neurons express c-FOS in response to light exposure in early night. Blockade of signals that relay light information, via NMDA receptors or VIP- and GRP-cognate receptors, has no effect on phase delays of circadian rhythms induced by little SAAS. Conclusions/Significance Little SAAS relays signals downstream of light/glutamatergic signaling from eye to SCN, and independent of VIP and GRP action. These findings suggest that little SAAS forms a third SCN neuropeptidergic system, processing light information and activating phase-shifts within novel circuits of the central circadian clock.


Journal of Proteome Research | 2013

Quantitative Peptidomics for Discovery of Circadian-Related Peptides from the Rat Suprachiasmatic Nucleus

Ji Eun Lee; Leonid Zamdborg; Bruce R. Southey; Norman Atkins; Jennifer W. Mitchell; Mingxi Li; Martha U. Gillette; Neil L. Kelleher; Jonathan V. Sweedler

In mammals the suprachiasmatic nucleus (SCN), the master circadian clock, is sensitive to light input via the optic chiasm and synchronizes many daily biological rhythms. Here we explore variations in the expression levels of neuropeptides present in the SCN of rats using a label-free quantification approach that is based on integrating peak intensities between daytime, Zeitgeber time (ZT) 6, and nighttime, ZT 18. From nine analyses comparing the levels between these two time points, 10 endogenous peptides derived from eight prohormones exhibited significant differences in their expression levels (adjusted p-value <0.05). Of these, seven peptides derived from six prohormones, including GRP, PACAP, and CART, exhibited ≥ 30% increases at ZT 18, and the VGRPEWWMDYQ peptide derived from proenkephalin A showed a >50% increase at nighttime. Several endogenous peptides showing statistically significant changes in this study have not been previously reported to alter their levels as a function of time of day, nor have they been implicated in prior functional SCN studies. This information on peptide expression changes serves as a resource for discovering unknown peptide regulators that affect circadian rhythms in the SCN.


Biochemical and Biophysical Research Communications | 2002

Synchronization and phase-resetting by glutamate of an immortalized SCN cell line.

William J. Hurst; Jennifer W. Mitchell; Martha U. Gillette

SCN 2.2 cultures were stably transfected with luciferase reporter constructs driven by Ca(2+)/cAMP response element, E-box, or vasoactive intestinal peptide promoter to probe the circadian properties of this clock cell line. SCN 2.2 reporter lines displayed approximately 24-h rhythms of transcriptional activation after serum-shock. Serum-shocked cultures pulsed with glutamate exhibited phase-gated induction of phospho-CREB and of VIP, CRE, and E-box promoter activity. Glutamate-induced CRE promoter activity displayed restricted sensitivity to inhibitors of nitric oxide synthase and cGMP-dependent protein kinase. The temporal pattern of these sensitivities paralleled those of the SCN to light and glutamate during the night. Taken together, our data indicate that serum-shock can synchronize the circadian clock of SCN 2.2 cells to a state consistent with the day/night transition and, thus, establishes a temporal context for this cell line.


Analytical Chemistry | 2014

Comparing label-free quantitative peptidomics approaches to characterize diurnal variation of peptides in the rat suprachiasmatic nucleus

Bruce R. Southey; Ji Eun Lee; Leonid Zamdborg; Norman Atkins; Jennifer W. Mitchell; Mingxi Li; Martha U. Gillette; Neil L. Kelleher; Jonathan V. Sweedler

Mammalian circadian rhythm is maintained by the suprachiasmatic nucleus (SCN) via an intricate set of neuropeptides and other signaling molecules. In this work, peptidomic analyses from two times of day were examined to characterize variation in SCN peptides using three different label-free quantitation approaches: spectral count, spectra index and SIEVE. Of the 448 identified peptides, 207 peptides were analyzed by two label-free methods, spectral count and spectral index. There were 24 peptides with significant (adjusted p-value < 0.01) differential peptide abundances between daytime and nighttime, including multiple peptides derived from secretogranin II, cocaine and amphetamine regulated transcript, and proprotein convertase subtilisin/kexin type 1 inhibitor. Interestingly, more peptides were analyzable and had significantly different abundances between the two time points using the spectral count and spectral index methods than with a prior analysis using the SIEVE method with the same data. The results of this study reveal the importance of using the appropriate data analysis approaches for label-free relative quantitation of peptides. The detection of significant changes in so rich a set of neuropeptides reflects the dynamic nature of the SCN and the number of influences such as feeding behavior on circadian rhythm. Using spectral count and spectral index, peptide level changes are correlated to time of day, suggesting their key role in circadian function.


Biochimica et Biophysica Acta | 2002

Identification of the calcium channel α1E (Cav2.3) isoform expressed in atrial myocytes

Jennifer W. Mitchell; Janice K. Larsen; Philip M. Best

Antisense oligonucleotides targeting the calcium channel alpha 1E (Ca(v)2.3) subunit significantly inhibit the insulin-like growth factor-1 (IGF-1)-stimulated increase in low voltage-activated (LVA) (T-type) calcium current in cultured rat atrial myocytes [Proc. Natl. Acad. Sci. U.S.A. 94(1997) 14936]. As part of a continuing effort to understand the regulation of LVA current expression in the heart, we have identified the specific alpha 1E isoform that is expressed in atrial tissue. Through reverse transcription-polymerase chain reaction (RT-PCR), nine overlapping partial clones spanning the entire coding region of the cardiac alpha 1E mRNA were obtained. The predominate isoform in atrial tissue was identified and found to be highly homologous to the alpha 1E isoform previously isolated from kidney and the islets of Langerhans [Eur. J. Biochem. 257(1998) 274]. The expression of alpha 1E in the heart occurs specifically in cardiac myocytes and not in smooth muscle or fibroblasts as demonstrated by RT-PCR performed on isolated atrial myocytes and by in situ hybridization.


Analyst | 2012

A hyphenated optical trap capillary electrophoresis laser induced native fluorescence system for single-cell chemical analysis

Christine Cecala; Stanislav S. Rubakhin; Jennifer W. Mitchell; Martha U. Gillette; Jonathan V. Sweedler

Single-cell measurements allow a unique glimpse into cell-to-cell heterogeneity; even small changes in selected cells can have a profound impact on an organisms physiology. Here an integrated approach to single-cell chemical sampling and assay are described. Capillary electrophoresis (CE) with laser-induced native fluorescence (LINF) has the sensitivity to characterize natively fluorescent indoles and catechols within individual cells. While the separation and detection approaches are well established, the sampling and injection of individually selected cells requires new approaches. We describe an optimized system that interfaces a single-beam optical trap with CE and multichannel LINF detection. A cell is localized within the trap and then the capillary inlet is positioned near the cell using a computer-controlled micromanipulator. Hydrodynamic injection allows cell lysis to occur within the capillary inlet, followed by the CE separation and LINF detection. The use of multiple emission wavelengths allows improved analyte identification based on differences in analyte fluorescence emission profiles and migration time. The system enables injections of individual rat pinealocytes and quantification of their endogenous indoles, including serotonin, N-acetyl-serotonin, 5-hydroxyindole-3-acetic acid, tryptophol and others. The amounts detected in individual cells incubated in 5-hydroxytryptophan ranged from 10(-14) mol to 10(-16) mol, an order of magnitude higher than observed in untreated pinealocytes.


Frontiers in Neuroendocrinology | 2011

Direct cellular peptidomics of hypothalamic neurons

Jennifer W. Mitchell; Norman Atkins; Jonathan V. Sweedler; Martha U. Gillette

The chemical complexity of cell-to-cell communication has emerged as a fundamental challenge to understanding brain systems. This is certainly true for the hypothalamus, where neuropeptide signals are heterogeneous, localized and dynamic. Thus far, most hypothalamic peptidomic studies have centered on the entire structure; however, recent advances in collection strategies and analytical technologies have enabled direct, high-resolution peptidomic profiles focused on two regions of interest, the suprachiasmatic and supraoptic nuclei, including their sub-regions and individual cells. Suites of peptides now can be identified and probed for function. High spatial and analytical sensitivities reveal that discrete hypothalamic nuclei have distinct peptidomic signatures. Peptidomic discovery not only reveals unanticipated complexity, but also peptides previously unknown that act as key circuit components. Analysis of tissue releasates identifies peptides secreted into the extracellular environment and available for transmitting intercellular signals. Direct sampling techniques define peptide-releasate profiles in spatial, temporal and event-dependent patterns. These approaches are providing remarkable new insights into the complexity of neuropeptidergic cell-to-cell signaling central to neuroendocrine physiology.


PLOS ONE | 2016

Melatonin Signal Transduction Pathways Require E-Box-Mediated Transcription of Per1 and Per2 to Reset the SCN Clock at Dusk

Patty C. Kandalepas; Jennifer W. Mitchell; Martha U. Gillette

Melatonin is released from the pineal gland into the circulatory system at night in the absence of light, acting as “hormone of darkness” to the brain and body. Melatonin also can regulate circadian phasing of the suprachiasmatic nucleus (SCN). During the day-to-night transition, melatonin exposure advances intrinsic SCN neural activity rhythms via the melatonin type-2 (MT2) receptor and downstream activation of protein kinase C (PKC). The effects of melatonin on SCN phasing have not been linked to daily changes in the expression of core genes that constitute the molecular framework of the circadian clock. Using real-time RT-PCR, we found that melatonin induces an increase in the expression of two clock genes, Period 1 (Per1) and Period 2 (Per2). This effect occurs at CT 10, when melatonin advances SCN phase, but not at CT 6, when it does not. Using anti-sense oligodeoxynucleotides (α ODNs) to Per 1 and Per 2, as well as to E-box enhancer sequences in the promoters of these genes, we show that their specific induction is necessary for the phase-altering effects of melatonin on SCN neural activity rhythms in the rat. These effects of melatonin on Per1 and Per2 were mediated by PKC. This is unlike day-active non-photic signals that reset the SCN clock by non-PCK signal transduction mechanisms and by decreasing Per1 expression. Rather, this finding extends roles for Per1 and Per2, which are critical to photic phase-resetting, to a nonphotic zeitgeber, melatonin, and suggest that the regulation of these clock gene transcripts is required for clock resetting by diverse regulatory cues.

Collaboration


Dive into the Jennifer W. Mitchell's collaboration.

Top Co-Authors

Avatar

Shelley A. Tischkau

Southern Illinois University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christian Beaulé

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Daniel J. Morgan

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason R. Hickok

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

John E. Pintar

University of Medicine and Dentistry of New Jersey

View shared research outputs
Researchain Logo
Decentralizing Knowledge