Try a new search

Format these results:

Searched for:

person:balapb01

in-biosketch:yes

Total Results:

71


Distinct functions of endogenous cannabinoid system in alcohol abuse disorders

Basavarajappa, Balapal S; Joshi, Vikram; Shivakumar, Madhu; Subbanna, Shivakumar
Δ9 -tetrahydrocannabinol, the principal active component in Cannabis sativa extracts such as marijuana, participates in cell signalling by binding to cannabinoid CB1 and CB2 receptors on the cell surface. The CB1 receptors are present in both inhibitory and excitatory presynaptic terminals and the CB2 receptors are found in neuronal subpopulations in addition to microglial cells and astrocytes and are present in both presynaptic and postsynaptic terminals. Subsequent to the discovery of the endocannabinoid (eCB) system, studies have suggested that alcohol alters the eCB system and that this system plays a major role in the motivation to abuse alcohol. Preclinical studies have provided evidence that chronic alcohol consumption modulates eCBs and expression of CB1 receptors in brain addiction circuits. In addition, studies have further established the distinct function of the eCB system in the development of fetal alcohol spectrum disorders. This review provides a recent and comprehensive assessment of the literature related to the function of the eCB system in alcohol abuse disorders.
PMCID:6692638
PMID: 31265740
ISSN: 1476-5381
CID: 4142392

CB1R mediated HDAC-EGR1 pathway causes neurobehavioral defects in postnatal ethanol exposedmice [Meeting Abstract]

Basavarajappa, B S; Subbanna, S; Joshi, V; Shivakumar, M
Alcohol abuse during pregnancy exposes the fetal brain to alcohol and impairs brain maturation, leading to persistent neurobehavioral abnormalities, including cognitive decline, which together is known as fetal alcohol spectrumdisorder (FASD). However, the molecular mechanisms triggering these developmental deficits are poorly explained. In this study, we report that the binge-type ethanol exposure of P7 mice, which activates caspase-3, enhanced the histone deacetylase (HDAC) 1, HDAC2 and HDAC3 levels and reduced histone 3 (lysine 14, K14) and histone 4 (lysine 8, K8) acetylation in mature neurons [neuron-specific nuclear (NeuN) positive]. Ethanol exposure repressed early growth response 1 (Egr1) gene and protein expression. The repressed gene promoter region displayed differential HDACs, enhanced G9a, H3K14ac and histone 3 (lysine 9, K9) dimethylation enrichment. However, CREB-binding protein (CBP) enrichment was reduced at Egr1 promoter region. Inhibition of class 1 HDACs with trichostatin (TSA) before ethanol exposure, rescued H3K14ac and H4K8ac levels and prevented caspase-3 activation. Antagonism or null mutation of cannabinoid receptor type-1 (CB1R) before ethanol exposure, which inhibits caspase-3 activation, prevented H3K14ac and H4K8ac loss. TSA administration before ethanol exposure prevented ethanol-induced loss of Egr1 expression, restored epigenetic remodeling and neurobehavioral defects in adult mice. Together, these findings demonstrate that ethanol-activated CB1R regulates epigenetic/ gene expression mechanisms causing persistent neurobehavioral defects. CB1R/HDAC-mediated epigenetic remodeling disrupts gene expression and is a critical step in cognitive decline development in FASD, which is reversed by restoring histone acetylation in the brain
EMBASE:628238923
ISSN: 1530-0277
CID: 4024692

CB1R regulates CDK5 signaling and epigenetically controls Rac1 expression contributing to neurobehavioral abnormalities in mice postnatally exposed to ethanol

Joshi, Vikram; Subbanna, Shivakumar; Shivakumar, Madhu; Basavarajappa, Balapal S
Fetal alcohol spectrum disorders (FASD) represent a wide array of defects that arise from ethanol exposure during development. However, the underlying molecular mechanisms are limited. In the current report, we aimed to further evaluate the cannabinoid receptor type 1 (CB1R)-mediated mechanisms in a postnatal ethanol-exposed animal model. We report that the exposure of postnatal day 7 (P7) mice to ethanol generates p25, a CDK5-activating peptide, in a time- and CB1R-dependent manner in the hippocampus and neocortex brain regions. Pharmacological inhibition of CDK5 activity before ethanol exposure prevented accumulation of cleaved caspase-3 (CC3) and hyperphosphorylated tau (PHF1) (a marker for neurodegeneration) in neonatal mice and reversed cAMP response element-binding protein (CREB) activation and activity-regulated cytoskeleton-associated protein (Arc) expression. We also found that postnatal ethanol exposure caused a loss of RhoGTPase-related, Rac1, gene expression in a CB1R and CDK5 activity-dependent manner, which persisted to adulthood. Our epigenetic analysis of the Rac1 gene promoter suggested that persistent suppression of Rac1 expression is mediated by enhanced histone H3 lysine 9 dimethylation (H3K9me2), a repressive chromatin state, via G9a recruitment. The inhibition of CDK5/p25 activity before postnatal ethanol exposure rescued CREB activation, Arc, chromatin remodeling and Rac1 expression, spatial memory, and long-term potentiation (LTP) abnormalities in adult mice. Together, these findings propose that the postnatal ethanol-induced CB1R-mediated activation of CDK5 suppresses Arc and Rac1 expression in the mouse brain and is responsible for persistent synaptic plasticity and learning and memory defects in adult mice. This CB1R-mediated activation of CDK5 signaling during active synaptic development may slow down the maturation of synaptic circuits and may cause neurobehavioral defects, as found in this FASD animal model.
PMCID:6333777
PMID: 30143782
ISSN: 1740-634x
CID: 4142372

Potential Mechanisms Underlying the Deleterious Effects of Synthetic Cannabinoids Found in Spice/K2 Products

Basavarajappa, Balapal S; Subbanna, Shivakumar
The chief psychoactive constituent of many bioactive phytocannabinoids (Δ⁸-tetrahydrocannabinol, Δ⁸-THC) found in hemp, cannabis or marijuana plants are scientifically denoted by the Latin term, Cannabis sativa, acts on cell surface receptors. These receptors are ubiquitously expressed. To date, two cannabinoid receptors have been cloned and characterized. Cannabinoid receptor type 1 (CB1R) is found to serve as the archetype for cannabinoid action in the brain. They have attracted wide interest as the mediator of all psychoactive properties of exogenous and endogenous cannabinoids and they are abundantly expressed on most inhibitory and excitatory neurons. Recent evidence established that cannabinoid receptor type 2 (CB2R) is also expressed in the neurons at both presynaptic and postsynaptic terminals and are involved in neuropsychiatric effects. Distinct types of cells in many regions in the brain express CB2Rs and the cellular origin of CB2Rs that induce specific behavioral effects are emerging. To mimic the bliss effects of marijuana, synthetic cannabinoids (SCBs) have been sprayed onto plant material, and this plant material has been consequently packaged and sold under brand name "Spice" or "K2". These SCBs have been shown to maintain their affinity and functional activity for CB1R and CB2R and have been shown to cause severe harmful effects when compared to the effects of Δ⁸-THC. The present review discusses the potential brain mechanisms that are involved in the deleterious effects of SCBs.
PMCID:6357179
PMID: 30654473
ISSN: 2076-3425
CID: 4142382

Endocannabinoid System and Alcohol Abuse Disorders

Basavarajappa, Balapal S
Δ9-tetrahydrocannabinol (Δ9-THC), the primary active component in Cannabis sativa preparations such as hashish and marijuana, signals by binding to cell surface receptors. Two types of receptors have been cloned and characterized as cannabinoid (CB) receptors. CB1 receptors (CB1R) are ubiquitously present in the central nervous system (CNS) and are present in both inhibitory interneurons and excitatory neurons at the presynaptic terminal. CB2 receptors (CB2R) are demonstrated in microglial cells, astrocytes, and several neuron subpopulations and are present in both pre- and postsynaptic terminals. The majority of studies on these receptors have been conducted in the past two and half decades after the identification of the molecular constituents of the endocannabinoid (eCB) system that started with the characterization of CB1R. Subsequently, the seminal discovery was made, which suggested that alcohol (ethanol) alters the eCB system, thus establishing the contribution of the eCB system in the motivation to consume ethanol. Several preclinical studies have provided evidence that CB1R significantly contributes to the motivational and reinforcing properties of ethanol and that the chronic consumption of ethanol alters eCB transmitters and CB1R expression in the brain nuclei associated with addiction pathways. Additionally, recent seminal studies have further established the role of the eCB system in the development of ethanol-induced developmental disorders, such as fetal alcohol spectrum disorders (FASD). These results are augmented by in vitro and ex vivo studies, showing that acute and chronic treatment with ethanol produces physiologically relevant alterations in the function of the eCB system during development and in the adult stage. This chapter provides a current and comprehensive review of the literature concerning the role of the eCB system in alcohol abuse disorders (AUD).
PMID: 31332736
ISSN: 0065-2598
CID: 4142402

Activity-dependent Signaling and Epigenetic Abnormalities in Mice Exposed to Postnatal Ethanol

Subbanna, Shivakumar; Joshi, Vikram; Basavarajappa, Balapal S
Postnatal ethanol exposure has been shown to cause persistent defects in hippocampal synaptic plasticity and disrupt learning and memory processes. However, the mechanisms responsible for these abnormalities are less well studied. We evaluated the influence of postnatal ethanol exposure on several signaling and epigenetic changes and on expression of the activity-regulated cytoskeletal (Arc) protein in the hippocampus of adult offspring under baseline conditions and after a Y-maze spatial memory (SP) behavior (activity). Postnatal ethanol treatment impaired pCaMKIV and pCREB in naïve mice without affecting H4K8ac, H3K14ac and H3K9me2 levels. The Y-maze increased pCaMKIV, pCREB, H4K8ac and H3K14ac levels in saline-treated mice but not in ethanol-treated mice; while H3K9me2 levels were enhanced in ethanol-exposed animals compared to saline groups. Like previous observations, ethanol not only reduced Arc expression in naïve mice but also behaviorally induced Arc expression. ChIP results suggested that reduced H3K14ac and H4K8ac in the Arc gene promoter is because of impaired CBP, and increased H3K9me2 is due to the enhanced recruitment of G9a. The CB1R antagonist and a G9a/GLP inhibitor, which were shown to rescue postnatal ethanol-triggered synaptic plasticity and learning and memory deficits, were able to prevent the negative effects of ethanol on activity-dependent signaling, epigenetics and Arc expression. Together, these findings provide a molecular mechanism involving signaling and epigenetic cascades that collectively are responsible for the neurobehavioral deficits associated with an animal model of fetal alcohol spectrum disorders (FASD).
PMCID:6204293
PMID: 30031835
ISSN: 1873-7544
CID: 4142362

Neurofilament light interaction with GluN1 modulates neurotransmission and schizophrenia-associated behaviors

Yuan, Aidong; Sershen, Henry; Basavarajappa, Balapal S; Smiley, John F; Hashim, Audrey; Bleiwas, Cynthia; Berg, Martin; Guifoyle, David N; Subbanna, Shivakumar; Darji, Sandipkumar; Kumar, Asok; Rao, Mala V; Wilson, Donald A; Julien, Jean-Pierre; Javitt, Daniel C; Nixon, Ralph A
Neurofilament (NFL) proteins have recently been found to play unique roles in synapses. NFL is known to interact with the GluN1 subunit of N-methyl-D-aspartic acid (NMDAR) and be reduced in schizophrenia though functional consequences are unknown. Here we investigated whether the interaction of NFL with GluN1 modulates synaptic transmission and schizophrenia-associated behaviors. The interaction of NFL with GluN1 was assessed by means of molecular, pharmacological, electrophysiological, magnetic resonance spectroscopy (MRS), and schizophrenia-associated behavior analyses. NFL deficits cause an NMDAR hypofunction phenotype including abnormal hippocampal function, as seen in schizophrenia. NFL-/- deletion in mice reduces dendritic spines and GluN1 protein levels, elevates ubiquitin-dependent turnover of GluN1 and hippocampal glutamate measured by MRS, and depresses hippocampal long-term potentiation. NMDAR-related behaviors are also impaired, including pup retrieval, spatial and social memory, prepulse inhibition, night-time activity, and response to NMDAR antagonist, whereas motor deficits are minimal. Importantly, partially lowering NFL in NFL+/- mice to levels seen regionally in schizophrenia, induced similar but milder NMDAR-related synaptic and behavioral deficits. Our findings support an emerging view that central nervous system neurofilament subunits including NFL in the present report, serve distinctive, critical roles in synapses relevant to neuropsychiatric diseases.
PMCID:6109052
PMID: 30143609
ISSN: 2158-3188
CID: 3246612

CB1R-Mediated Activation of Caspase-3 Causes Epigenetic and Neurobehavioral Abnormalities in Postnatal Ethanol-Exposed Mice

Subbanna, Shivakumar; Nagre, Nagaraja N; Shivakumar, Madhu; Joshi, Vikram; Psychoyos, Delphine; Kutlar, Abdullah; Umapathy, Nagavedi S; Basavarajappa, Balapal S
Alcohol exposure can affect brain development, leading to long-lasting behavioral problems, including cognitive impairment, which together is defined as fetal alcohol spectrum disorder (FASD). However, the fundamental mechanisms through which this occurs are largely unknown. In this study, we report that the exposure of postnatal day 7 (P7) mice to ethanol activates caspase-3 via cannabinoid receptor type-1 (CB1R) in neonatal mice and causes a reduction in methylated DNA binding protein (MeCP2) levels. The developmental expression of MeCP2 in mice is closely correlated with synaptogenesis and neuronal maturation. It was shown that ethanol treatment of P7 mice enhanced Mecp2 mRNA levels but reduced protein levels. The genetic deletion of CB1R prevented, and administration of a CB1R antagonist before ethanol treatment of P7 mice inhibited caspase-3 activation. Additionally, it reversed the loss of MeCP2 protein, cAMP response element binding protein (CREB) activation, and activity-regulated cytoskeleton-associated protein (Arc) expression. The inhibition of caspase-3 activity prior to ethanol administration prevented ethanol-induced loss of MeCP2, CREB activation, epigenetic regulation of Arc expression, long-term potentiation (LTP), spatial memory deficits and activity-dependent impairment of several signaling molecules, including MeCP2, in adult mice. Collectively, these results reveal that the ethanol-induced CB1R-mediated activation of caspase-3 degrades the MeCP2 protein in the P7 mouse brain and causes long-lasting neurobehavioral deficits in adult mice. This CB1R-mediated instability of MeCP2 during active synaptic maturation may disrupt synaptic circuit maturation and lead to neurobehavioral abnormalities, as observed in this animal model of FASD.
PMCID:5826222
PMID: 29515368
ISSN: 1662-5099
CID: 4142352

Endocannabinoid system in neurodegenerative disorders

Basavarajappa, Balapal S; Shivakumar, Madhu; Joshi, Vikram; Subbanna, Shivakumar
Most neurodegenerative disorders (NDDs) are characterized by cognitive impairment and other neurological defects. The definite cause of and pathways underlying the progression of these NDDs are not well-defined. Several mechanisms have been proposed to contribute to the development of NDDs. These mechanisms may proceed concurrently or successively, and they differ among cell types at different developmental stages in distinct brain regions. The endocannabinoid system, which involves cannabinoid receptors type 1 (CB1R) and type 2 (CB2R), endogenous cannabinoids and the enzymes that catabolize these compounds, has been shown to contribute to the development of NDDs in several animal models and human studies. In this review, we discuss the functions of the endocannabinoid system in NDDs and converse the therapeutic efficacy of targeting the endocannabinoid system to rescue NDDs.
PMCID:5669051
PMID: 28608560
ISSN: 1471-4159
CID: 4142342

Cannabinoid receptors and their signaling mechanisms

Chapter by: Basavarajappa, Balapal S
in: The endocannabinoid system by Murillo-Rodriguez, Eric (Ed)
London, United Kingdom ; San Diego, CA : Academic Press, [2017]
pp. 25-62
ISBN: 0128096667
CID: 4142452