Network


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

Hotspot


Dive into the research topics where Karen H. Berger is active.

Publication


Featured researches published by Karen H. Berger.


Cell | 2009

Happyhour, a Ste20 Family Kinase, Implicates EGFR Signaling in Ethanol-Induced Behaviors

Ammon B. Corl; Karen H. Berger; Julie Gesch; Jeffrey A. Simms; Selena E. Bartlett; Ulrike Heberlein

The consequences of alcohol use disorders (AUDs) are devastating to individuals and society, yet few treatments are currently available. To identify genes regulating the behavioral effects of ethanol, we conducted a genetic screen in Drosophila and identified a mutant, happyhour (hppy), due to its increased resistance to the sedative effects of ethanol. Hppy protein shows strong homology to mammalian Ste20 family kinases of the GCK-1 subfamily. Genetic and biochemical experiments revealed that the epidermal growth factor (EGF)-signaling pathway regulates ethanol sensitivity in Drosophila and that Hppy functions as an inhibitor of the pathway. Acute pharmacological inhibition of the EGF receptor (EGFR) in adult animals altered acute ethanol sensitivity in both flies and mice and reduced ethanol consumption in a preclinical rat model of alcoholism. Inhibitors of the EGFR or components of its signaling pathway are thus potential pharmacotherapies for AUDs.


PLOS ONE | 2011

An Evolutionary Conserved Role for Anaplastic Lymphoma Kinase in Behavioral Responses to Ethanol

Amy W. Lasek; Jana Lim; Christopher L. Kliethermes; Karen H. Berger; Geoff Joslyn; Gerry Brush; Liquan Xue; Margaret Robertson; Monica S. Moore; Karen Vranizan; Stephan W. Morris; Marc A. Schuckit; Raymond L. White; Ulrike Heberlein

Anaplastic lymphoma kinase (Alk) is a gene expressed in the nervous system that encodes a receptor tyrosine kinase commonly known for its oncogenic function in various human cancers. We have determined that Alk is associated with altered behavioral responses to ethanol in the fruit fly Drosophila melanogaster, in mice, and in humans. Mutant flies containing transposon insertions in dAlk demonstrate increased resistance to the sedating effect of ethanol. Database analyses revealed that Alk expression levels in the brains of recombinant inbred mice are negatively correlated with ethanol-induced ataxia and ethanol consumption. We therefore tested Alk gene knockout mice and found that they sedate longer in response to high doses of ethanol and consume more ethanol than wild-type mice. Finally, sequencing of human ALK led to the discovery of four polymorphisms associated with a low level of response to ethanol, an intermediate phenotype that is predictive of future alcohol use disorders (AUDs). These results suggest that Alk plays an evolutionary conserved role in ethanol-related behaviors. Moreover, ALK may be a novel candidate gene conferring risk for AUDs as well as a potential target for pharmacological intervention.


Alcoholism: Clinical and Experimental Research | 2004

Rapid and chronic: two distinct forms of ethanol tolerance in Drosophila.

Karen H. Berger; Ulrike Heberlein; Monica S. Moore

BACKGROUND Ethanol tolerance, defined as a reduction in the intensity of the effects of ethanol upon continuous or repeated exposure, is a hallmark of alcoholism. Tolerance may develop at the cellular or neural systems levels. The molecular changes underlying ethanol tolerance are not well understood. We therefore explored the utility of Drosophila, with its accessibility to genetic, molecular, and behavioral analyses, as a model organism to study tolerance development in response to different ethanol-exposure regimens. METHODS We describe a new assay that quantifies recovery from ethanol intoxication in Drosophila. Using this recovery assay, we define ethanol pre-exposure paradigms that lead to the development of tolerance. We also use the inebriometer, an assay that measures the onset of intoxication, to study the effects of pharmacological and genetic manipulations on tolerance development. RESULTS We show that flies develop different forms of ethanol tolerance: rapid tolerance, induced by a single short exposure to a high concentration of ethanol, and chronic tolerance, elicited by prolonged exposure to a low concentration of the drug. Neither rapid nor chronic tolerance involves changes in ethanol pharmacokinetics, implying that they represent functional rather than dispositional tolerance. Chronic and rapid tolerance can be distinguished mechanistically: chronic tolerance is disrupted by treatment with the protein synthesis inhibitor cycloheximide, whereas rapid tolerance is resistant to this treatment. Furthermore, rapid and chronic tolerance rely on distinct genetic pathways: a mutant defective for octopamine biosynthesis shows reduced rapid tolerance but normal chronic tolerance. CONCLUSIONS Flies, like mammals, develop tolerance in response to different ethanol-exposure regimens, and this tolerance affects both the onset of and the recovery from acute intoxication. Two forms of tolerance, rapid and chronic, are mechanistically distinct, because they can be dissociated genetically and pharmacologically.


Alcoholism: Clinical and Experimental Research | 2008

Ethanol Sensitivity and Tolerance in Long-Term Memory Mutants of Drosophila melanogaster

Karen H. Berger; Eric C. Kong; Josh Dubnau; Tim Tully; Monica S. Moore; Ulrike Heberlein

BACKGROUND It has become increasingly clear that molecular and neural mechanisms underlying learning and memory and drug addiction are largely shared. To confirm and extend these findings, we analyzed ethanol-responsive behaviors of a collection of Drosophila long-term memory mutants. METHODS For each mutant, sensitivity to the acute uncoordinating effects of ethanol was quantified using the inebriometer. Additionally, 2 distinct forms of ethanol tolerance were measured: rapid tolerance, which develops in response to a single brief exposure to a high concentration of ethanol vapor; and chronic tolerance, which develops following a sustained low-level exposure. RESULTS Several mutants were identified with altered sensitivity, rapid or chronic tolerance, while a number of mutants exhibited multiple defects. CONCLUSIONS The corresponding genes in these mutants represent areas of potential overlap between learning and memory and behavioral responses to alcohol. These genes also define components shared between different ethanol behavioral responses.


The Journal of Neuroscience | 2010

Protein Phosphatase 2A and Glycogen Synthase Kinase 3 Signaling Modulate Prepulse Inhibition of the Acoustic Startle Response by Altering Cortical M-Type Potassium Channel Activity

David Kapfhamer; Karen H. Berger; F. Woodward Hopf; Taban Seif; Viktor Kharazia; Antonello Bonci; Ulrike Heberlein

There is considerable interest in the regulation of sensorimotor gating, since deficits in this process could play a critical role in the symptoms of schizophrenia and other psychiatric disorders. Sensorimotor gating is often studied in humans and rodents using the prepulse inhibition of the acoustic startle response (PPI) model, in which an acoustic prepulse suppresses behavioral output to a startle-inducing stimulus. However, the molecular and neural mechanisms underlying PPI are poorly understood. Here, we show that a regulatory pathway involving protein phosphatase 2A (PP2A), glycogen synthase kinase 3 β (GSK3β), and their downstream target, the M-type potassium channel, regulates PPI. Mice (Mus musculus) carrying a hypomorphic allele of Ppp2r5δ, encoding a regulatory subunit of PP2A, show attenuated PPI. This PPP2R5δ reduction increases the phosphorylation of GSK3β at serine 9, which inactivates GSK3β, indicating that PPP2R5δ positively regulates GSK3β activity in the brain. Consistently, genetic and pharmacological manipulations that reduce GSK3β function attenuate PPI. The M-type potassium channel subunit, KCNQ2, is a putative GSK3β substrate. Genetic reduction of Kcnq2 also reduces PPI, as does systemic inhibition of M-channels with linopirdine. Importantly, both the GSK3 inhibitor 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)1H-pyrrole-2,5-dione (SB216763) and linopirdine reduce PPI when directly infused into the medial prefrontal cortex (mPFC). Whole-cell electrophysiological recordings of mPFC neurons show that SB216763 and linopirdine have similar effects on firing, and GSK3 inhibition occludes the effects of M-channel inhibition. These data support a previously uncharacterized mechanism by which PP2A/GSK3β signaling regulates M-type potassium channel activity in the mPFC to modulate sensorimotor gating.


Neurotoxicology | 2009

Genetic aspects of behavioral neurotoxicology.

Edward D. Levin; Michael Aschner; Ulrike Heberlein; Douglas M. Ruden; Kathleen A. Welsh-Bohmer; Selena E. Bartlett; Karen H. Berger; Lang Chen; Ammon B. Corl; Donnie Eddins; Rachael L. French; Kathleen M. Hayden; Kirsten J. Helmcke; Helmut V. B. Hirsch; Elwood Linney; Greg Lnenicka; Grier P. Page; Debra Possidente; Bernard Possidente; Annette Kirshner

Considerable progress has been made over the past couple of decades concerning the molecular bases of neurobehavioral function and dysfunction. The field of neurobehavioral genetics is becoming mature. Genetic factors contributing to neurologic diseases such as Alzheimers disease have been found and evidence for genetic factors contributing to other diseases such as schizophrenia and autism are likely. This genetic approach can also benefit the field of behavioral neurotoxicology. It is clear that there is substantial heterogeneity of response with behavioral impairments resulting from neurotoxicants. Many factors contribute to differential sensitivity, but it is likely that genetic variability plays a prominent role. Important discoveries concerning genetics and behavioral neurotoxicity are being made on a broad front from work with invertebrate and piscine mutant models to classic mouse knockout models and human epidemiologic studies of polymorphisms. Discovering genetic factors of susceptibility to neurobehavioral toxicity not only helps identify those at special risk, it also advances our understanding of the mechanisms by which toxicants impair neurobehavioral function in the larger population. This symposium organized by Edward Levin and Annette Kirshner, brought together researchers from the laboratories of Michael Aschner, Douglas Ruden, Ulrike Heberlein, Edward Levin and Kathleen Welsh-Bohmer conducting studies with Caenorhabditis elegans, Drosophila, fish, rodents and humans studies to determine the role of genetic factors in susceptibility to behavioral impairment from neurotoxic exposure.


PLOS ONE | 2012

DlgS97/SAP97, a Neuronal Isoform of Discs Large, Regulates Ethanol Tolerance

Rajani Maiya; Seonok Lee; Karen H. Berger; Eric C. Kong; Justin B. Slawson; Leslie C. Griffith; Kogo Takamiya; Richard L. Huganir; Ben Margolis; Ulrike Heberlein

From a genetic screen for Drosophila melanogaster mutants with altered ethanol tolerance, we identified intolerant (intol), a novel allele of discs large 1 (dlg1). Dlg1 encodes Discs Large 1, a MAGUK (Membrane Associated Guanylate Kinase) family member that is the highly conserved homolog of mammalian PSD-95 and SAP97. The intol mutation disrupted specifically the expression of DlgS97, a SAP97 homolog, and one of two major protein isoforms encoded by dlg1 via alternative splicing. Expression of the major isoform, DlgA, a PSD-95 homolog, appeared unaffected. Ethanol tolerance in the intol mutant could be partially restored by transgenic expression of DlgS97, but not DlgA, in specific neurons of the fly’s brain. Based on co-immunoprecipitation, DlgS97 forms a complex with N-methyl-D-aspartate (NMDA) receptors, a known target of ethanol. Consistent with these observations, flies expressing reduced levels of the essential NMDA receptor subunit dNR1 also showed reduced ethanol tolerance, as did mutants in the gene calcium/calmodulin-dependent protein kinase (caki), encoding the fly homolog of mammalian CASK, a known binding partner of DlgS97. Lastly, mice in which SAP97, the mammalian homolog of DlgS97, was conditionally deleted in adults failed to develop rapid tolerance to ethanol’s sedative/hypnotic effects. We propose that DlgS97/SAP97 plays an important and conserved role in the development of tolerance to ethanol via NMDA receptor-mediated synaptic plasticity.


Alcoholism: Clinical and Experimental Research | 2011

Lmo Genes Regulate Behavioral Responses to Ethanol in Drosophila melanogaster and the Mouse

Amy W. Lasek; Francesco Giorgetti; Karen H. Berger; Stacy Tayor; Ulrike Heberlein


Archive | 2006

Ethanol-responsive behaviors & learning and memory-shared mechanisms?

Karen H. Berger; Eric C. Kong; Josh Dubnau; Tim Tully; Monica S. Moore; Ulrike Heberlein


Alcoholism: Clinical and Experimental Research | 2005

Letters to the editor (multiple letters)

Svetlana Dzitoyeva; Nikola Dimitrijevic; Hari Manev; Karen H. Berger; Monica S. Moore; Ulrike Heberlein

Collaboration


Dive into the Karen H. Berger's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric C. Kong

University of California

View shared research outputs
Top Co-Authors

Avatar

Ammon B. Corl

University of California

View shared research outputs
Top Co-Authors

Avatar

Amy W. Lasek

University of California

View shared research outputs
Top Co-Authors

Avatar

Josh Dubnau

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Tim Tully

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Selena E. Bartlett

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Annette Kirshner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Antonello Bonci

National Institute on Drug Abuse

View shared research outputs
Researchain Logo
Decentralizing Knowledge