Network


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

Hotspot


Dive into the research topics where Marlin H. Dehoff is active.

Publication


Featured researches published by Marlin H. Dehoff.


Neuron | 2004

Homer Proteins Regulate Sensitivity to Cocaine

Karen K. Szumlinski; Marlin H. Dehoff; Shin H. Kang; Kelly A. Frys; Kevin D. Lominac; Matthias Klugmann; Jason Rohrer; William C. Griffin; Shigenobu Toda; Nicolas P. Champtiaux; Thomas L. Berry; Jian C. Tu; Stephanie E. Shealy; Matthew J. During; Lawrence D. Middaugh; Paul F. Worley; Peter W. Kalivas

Drug addiction involves complex interactions between pharmacology and learning in genetically susceptible individuals. Members of the Homer gene family are regulated by acute and chronic cocaine administration. Here, we report that deletion of Homer1 or Homer2 in mice caused the same increase in sensitivity to cocaine-induced locomotion, conditioned reward, and augmented extracellular glutamate in nucleus accumbens as that elicited by withdrawal from repeated cocaine administration. Moreover, adeno-associated virus-mediated restoration of Homer2 in the accumbens of Homer2 KO mice reversed the cocaine-sensitized phenotype. Further analysis of Homer2 KO mice revealed extensive additional behavioral and neurochemical similarities to cocaine-sensitized animals, including accelerated acquisition of cocaine self-administration and altered regulation of glutamate by metabotropic glutamate receptors and cystine/glutamate exchange. These data show that Homer deletion mimics the behavioral and neurochemical phenotype produced by repeated cocaine administration and implicate Homer in regulating addiction to cocaine.


The Journal of Neuroscience | 2005

Homer2 Is Necessary for EtOH-Induced Neuroplasticity

Karen K. Szumlinski; Kevin D. Lominac; Erik B. Oleson; Jennifer K. Walker; Ashley R. Mason; Marlin H. Dehoff; Matthias Klugman; Stephanie Cagle; Kristine Welt; Matthew J. During; Paul F. Worley; Lawrence D. Middaugh; Peter W. Kalivas

Homer proteins are integral to the assembly of proteins regulating glutamate signaling and synaptic plasticity. Constitutive Homer2 gene deletion [knock-out (KO)] and rescue with adeno-associated viral (AAV) transfection of Homer2b was used to demonstrate the importance of Homer proteins in neuroplasticity produced by repeated ethanol (EtOH) administration. Homer2 KO mice avoided drinking high concentrations of EtOH and did not develop place preference or locomotor sensitization after repeated EtOH administration. The deficient behavioral plasticity to EtOH after Homer2 deletion was paralleled by a lack of augmentation in the rise in extracellular dopamine and glutamate elicited by repeated EtOH injections. The genotypic differences in EtOH-induced change in behavior and neurochemistry were essentially reversed by AAV-mediated transfection of Homer2b into accumbens cells including, differences in EtOH preference, locomotor sensitization, and EtOH-induced elevations in extracellular glutamate and dopamine. These data demonstrate a necessary and active role for accumbens Homer2 expression in regulating EtOH-induced behavioral and cellular neuroplasticity.


Neuron | 2010

Homeostatic Scaling Requires Group I mGluR Activation Mediated by Homer1a

Jia Hua Hu; Joo Min Park; Sungjin Park; Bo Xiao; Marlin H. Dehoff; Sangmok Kim; Takashi Hayashi; Martin K. Schwarz; Richard L. Huganir; Peter H. Seeburg; David J. Linden; Paul F. Worley

Homeostatic scaling is a non-Hebbian form of neural plasticity that maintains neuronal excitability and informational content of synaptic arrays in the face of changes of network activity. Here, we demonstrate that homeostatic scaling is dependent on group I metabotropic glutamate receptor activation that is mediated by the immediate early gene Homer1a. Homer1a is transiently upregulated during increases in network activity and evokes agonist-independent signaling of group I mGluRs that scales down the expression of synaptic AMPA receptors. Homer1a effects are dynamic and play a role in the induction of scaling. Similar to mGluR-LTD, Homer1a-dependent scaling involves a reduction of tyrosine phosphorylation of GluA2 (GluR2), but is distinct in that it exploits a unique signaling property of group I mGluR to confer cell-wide, agonist-independent activation of the receptor. These studies reveal an elegant interplay of mechanisms that underlie Hebbian and non-Hebbian plasticity.


Genes, Brain and Behavior | 2005

Behavioral and neurochemical phenotyping of Homer1 mutant mice: possible relevance to schizophrenia

Karen K. Szumlinski; Kevin D. Lominac; M. J. Kleschen; Erik B. Oleson; Marlin H. Dehoff; M. K. Schwartz; P. H. Seeberg; Paul F. Worley; Peter W. Kalivas

Homer proteins are involved in the functional assembly of postsynaptic density proteins at glutamatergic synapses and are implicated in learning, memory and drug addiction. Here, we report that Homer1‐knockout (Homer1‐KO) mice exhibit behavioral and neurochemical abnormalities that are consistent with the animal models of schizophrenia. Relative to wild‐type mice, Homer1‐KO mice exhibited deficits in radial arm maze performance, impaired prepulse inhibition, enhanced ‘behavioral despair’, increased anxiety in a novel objects test, enhanced reactivity to novel environments, decreased instrumental responding for sucrose and enhanced MK‐801‐ and methamphetamine‐stimulated motor behavior. No‐net‐flux in vivo microdialysis revealed a decrease in extracellular glutamate content in the nucleus accumbens and an increase in the prefrontal cortex. Moreover, in Homer1‐KO mice, cocaine did not stimulate a rise in frontal cortex extracellular glutamate levels, suggesting hypofrontality. These behavioral and neurochemical data derived from Homer1 mutant mice are consistent with the recent association of schizophrenia with a single‐nucleotide polymorphism in the Homer1 gene and suggest that the regulation of extracellular levels of glutamate within limbo‐corticostriatal structures by Homer1 gene products may be involved in the pathogenesis of this neuropsychiatric disorder.


Cell | 2011

Arc/Arg3.1 Regulates an Endosomal Pathway Essential for Activity-Dependent β-Amyloid Generation

Jing Wu; Ronald S. Petralia; Hideaki Kurushima; Hiral Patel; Mi Young Jung; Lenora Volk; Shoaib Chowdhury; Jason D. Shepherd; Marlin H. Dehoff; Yueming Li; Dietmar Kuhl; Richard L. Huganir; Donald L. Price; Robert H. Scannevin; Juan C. Troncoso; Philip C. Wong; Paul F. Worley

Assemblies of β-amyloid (Aβ) peptides are pathological mediators of Alzheimers Disease (AD) and are produced by the sequential cleavages of amyloid precursor protein (APP) by β-secretase (BACE1) and γ-secretase. The generation of Aβ is coupled to neuronal activity, but the molecular basis is unknown. Here, we report that the immediate early gene Arc is required for activity-dependent generation of Aβ. Arc is a postsynaptic protein that recruits endophilin2/3 and dynamin to early/recycling endosomes that traffic AMPA receptors to reduce synaptic strength in both hebbian and non-hebbian forms of plasticity. The Arc-endosome also traffics APP and BACE1, and Arc physically associates with presenilin1 (PS1) to regulate γ-secretase trafficking and confer activity dependence. Genetic deletion of Arc reduces Aβ load in a transgenic mouse model of AD. In concert with the finding that patients with AD can express anomalously high levels of Arc, we hypothesize that Arc participates in the pathogenesis of AD.


The Journal of Neuroscience | 2009

Binge Drinking Upregulates Accumbens mGluR5–Homer2–PI3K Signaling: Functional Implications for Alcoholism

Debra K. Cozzoli; Scott P. Goulding; Ping Wu Zhang; Bo Xiao; Jia-Hua Hu; Alexis W. Ary; Ilona Obara; Alison Rahn; Hoda Abou-Ziab; Burgundy Tyrrel; Christina Marini; Naomi Yoneyama; Pamela Metten; Christopher Snelling; Marlin H. Dehoff; John C. Crabbe; Deborah A. Finn; Matthias Klugmann; Paul F. Worley; Karen K. Szumlinski

The glutamate receptor-associated protein Homer2 regulates alcohol-induced neuroplasticity within the nucleus accumbens (NAC), but the precise intracellular signaling cascades involved are not known. This study examined the role for NAC metabotropic glutamate receptor (mGluR)–Homer2–phosphatidylinositol 3-kinase (PI3K) signaling in regulating excessive alcohol consumption within the context of the scheduled high alcohol consumption (SHAC) model of binge alcohol drinking. Repeated bouts of binge drinking (∼1.5 g/kg per 30 min) elevated NAC Homer2a/b expression and increased PI3K activity in this region. Virus-mediated knockdown of NAC Homer2b expression attenuated alcohol intake, as did an intra-NAC infusion of the mGluR5 antagonist MPEP [2-methyl-6-(phenylethynyl)pyridine hydrochloride] (0.1–1 μg/side) and the PI3K antagonist wortmannin (50 ng/side), supporting necessary roles for mGluR5/Homer2/PI3K in binge alcohol drinking. Moreover, when compared with wild-type littermates, transgenic mice with an F1128R point mutation in mGluR5 that markedly reduces Homer binding exhibited a 50% reduction in binge alcohol drinking, which was related to reduced NAC basal PI3K activity. Consistent with the hypothesis that mGluR5–Homer–PI3K signaling may be a mechanism governing excessive alcohol intake, the “anti-binge” effects of MPEP and wortmannin were not additive, nor were they observed in the mGluR5F1128R transgenic mice. Finally, mice genetically selected for a high versus low SHAC phenotype differed in NAC mGluR, Homer2, and PI3K activity, consistent with the hypothesis that augmented NAC mGluR5–Homer2–PI3K signaling predisposes a high binge alcohol-drinking phenotype. Together, these data point to an important role for NAC mGluR5–Homer2–PI3K signaling in regulating binge-like alcohol consumption that has relevance for our understanding of the neurobiology of alcoholism and its pharmacotherapy.


Journal of Biological Chemistry | 2006

Homer 1 mediates store- and inositol 1,4,5-trisphosphate receptor-dependent translocation and retrieval of TRPC3 to the plasma membrane.

Joo Young Kim; Weizong Zeng; Kirill Kiselyov; Joseph P. Yuan; Marlin H. Dehoff; Katsuhiko Mikoshiba; Paul F. Worley; Shmuel Muallem

Store-operated Ca2+ channels (SOCs) mediate receptor-stimulated Ca2+ influx. Accumulating evidence indicates that members of the transient receptor potential (TRP) channel family are components of SOCs in mammalian cells. Agonist stimulation activates SOCs and TRP channels directly and by inducing translocation of channels in intracellular vesicles to the plasma membrane (PM). The mechanism of TRP channel translocation in response to store depletion and agonist stimulation is not known. Here we use TRPC3 as a model to show that IP3 and the scaffold Homer 1 (H1) regulate the rate of translocation and retrieval of TRPC3 from the PM. In resting cells, TRPC3 exists in TRPC3-H1b/c-IP3Rs complexes that are located in part at the PM and in part in intracellular vesicles. Binding of IP3 to the IP3Rs dissociates the interaction between IP3Rs and H1 but not between H1 and TRPC3 to form IP3Rs-TRPC3-H1b/c. TIRFM and biotinylation assays show robust receptor- and store-dependent translocation of the TRPC3 to the PM and their retrieval upon termination of cell stimulation. The translocation requires depletion of stored Ca2+ and is prevented by inhibition of the IP3Rs. In HEK293, dissociating the H1b/c-IP3R complex with H1a results in TRPC3 translocation to the PM, where it is spontaneously active. The TRPC3-H1b/c-IP3Rs complex is reconstituted by infusing H1c into these cells. Reconstitution is inhibited by IP3. Deletion of H1 in mice markedly reduces the rates of translocation and retrieval of TRPC3. Conversely, infusion of H1c into H1-/- cells eliminates spontaneous channel activity and increases the rate of channel activation by agonist stimulation. The effects of H1c are inhibited by IP3. These findings together with our earlier studies demonstrating gating of TRPC3 by IP3Rs were used to develop a model in which assembly of the TRPC3-H1b/c-IP3Rs complexes by H1b/c mediates both the translocation of TRPC3-containing vesicles to the PM and gating of TRPC3 by IP3Rs.


Journal of Cell Biology | 2003

Homer 2 tunes G protein–coupled receptors stimulus intensity by regulating RGS proteins and PLCβ GAP activities

Dong Min Shin; Marlin H. Dehoff; Xiang Luo; Shin Hyeok Kang; Jiangchen Tu; Surendra K. Nayak; Elliott M. Ross; Paul F. Worley; Shmuel Muallem

Homers are scaffolding proteins that bind G protein–coupled receptors (GPCRs), inositol 1,4,5-triphosphate (IP3) receptors (IP3Rs), ryanodine receptors, and TRP channels. However, their role in Ca2+ signaling in vivo is not known. Characterization of Ca2+ signaling in pancreatic acinar cells from Homer2−/− and Homer3−/− mice showed that Homer 3 has no discernible role in Ca2+ signaling in these cells. In contrast, we found that Homer 2 tunes intensity of Ca2+ signaling by GPCRs to regulate the frequency of [Ca2+]i oscillations. Thus, deletion of Homer 2 increased stimulus intensity by increasing the potency for agonists acting on various GPCRs to activate PLCβ and evoke Ca2+ release and oscillations. This was not due to aberrant localization of IP3Rs in cellular microdomains or IP3R channel activity. Rather, deletion of Homer 2 reduced the effectiveness of exogenous regulators of G proteins signaling proteins (RGS) to inhibit Ca2+ signaling in vivo. Moreover, Homer 2 preferentially bound to PLCβ in pancreatic acini and brain extracts and stimulated GAP activity of RGS4 and of PLCβ in an in vitro reconstitution system, with minimal effect on PLCβ-mediated PIP2 hydrolysis. These findings describe a novel, unexpected function of Homer proteins, demonstrate that RGS proteins and PLCβ GAP activities are regulated functions, and provide a molecular mechanism for tuning signal intensity generated by GPCRs and, thus, the characteristics of [Ca2+]i oscillations.


Science | 2008

NFAT Binding and Regulation of T Cell Activation by the Cytoplasmic Scaffolding Homer Proteins

Guo N. Huang; David L. Huso; Samuel Bouyain; Jianchen Tu; Kelly A. McCorkell; Michael J. May; Yuwen Zhu; Michael A. Lutz; Samuel L. Collins; Marlin H. Dehoff; Shin Kang; Katharine A. Whartenby; Jonathan D. Powell; Daniel J. Leahy; Paul F. Worley

T cell receptor (TCR) and costimulatory receptor (CD28) signals cooperate in activating T cells, although understanding of how these pathways are themselves regulated is incomplete. We found that Homer2 and Homer3, members of the Homer family of cytoplasmic scaffolding proteins, are negative regulators of T cell activation. This is achieved through binding of nuclear factor of activated T cells (NFAT) and by competing with calcineurin. Homer-NFAT binding was also antagonized by active serine-threonine kinase AKT, thereby enhancing TCR signaling via calcineurin-dependent dephosphorylation of NFAT. This corresponded with changes in cytokine expression and an increase in effector-memory T cell populations in Homer-deficient mice, which also developed autoimmune-like pathology. These results demonstrate a further means by which costimulatory signals are regulated to control self-reactivity.


Genes, Brain and Behavior | 2007

Complex, multimodal behavioral profile of the Homer1 knockout mouse

P. J. Jaubert; Mari S. Golub; Y. Y. Lo; S. L. Germann; Marlin H. Dehoff; Paul F. Worley; Shin H. Kang; Martin K. Schwarz; Peter H. Seeburg; Robert F. Berman

Proteins of the Homer1 immediate early gene family have been associated with synaptogenesis and synaptic plasticity suggesting broad behavioral consequences of loss of function. This study examined the behavior of male Homer1 knockout (KO) mice compared with wild‐type (WT) and heterozygous mice using a battery of 10 behavioral tests probing sensory, motor, social, emotional and learning/memory functions. KO mice showed mild somatic growth retardation, poor motor coordination, enhanced sensory reactivity and learning deficits. Heterozygous mice showed increased aggression in social interactions with conspecifics. The distribution of mGluR5 and N‐methyl‐d‐aspartate receptors (NMDA) receptors appeared to be unaltered in the hippocampus (HIP) of Homer1 KO mice. The results indicate an extensive range of disrupted behaviors that should contribute to the understanding of the Homer1 gene in brain development and behavior.

Collaboration


Dive into the Marlin H. Dehoff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter W. Kalivas

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Shmuel Muallem

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Xiao

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard L. Huganir

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge