Network


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

Hotspot


Dive into the research topics where Irina N. Krasnova is active.

Publication


Featured researches published by Irina N. Krasnova.


Brain Research Reviews | 2009

Methamphetamine toxicity and messengers of death

Irina N. Krasnova; Jean Lud Cadet

Methamphetamine (METH) is an illicit psychostimulant that is widely abused in the world. Several lines of evidence suggest that chronic METH abuse leads to neurodegenerative changes in the human brain. These include damage to dopamine and serotonin axons, loss of gray matter accompanied by hypertrophy of the white matter and microgliosis in different brain areas. In the present review, we summarize data on the animal models of METH neurotoxicity which include degeneration of monoaminergic terminals and neuronal apoptosis. In addition, we discuss molecular and cellular bases of METH-induced neuropathologies. The accumulated evidence indicates that multiple events, including oxidative stress, excitotoxicity, hyperthermia, neuroinflammatory responses, mitochondrial dysfunction, and endoplasmic reticulum stress converge to mediate METH-induced terminal degeneration and neuronal apoptosis. When taken together, these findings suggest that pharmacological strategies geared towards the prevention and treatment of the deleterious effects of this drug will need to attack the various pathways that form the substrates of METH toxicity.


Neurotoxicity Research | 2007

Neurotoxicity of substituted amphetamines: molecular and cellular mechanisms.

Jean Lud Cadet; Irina N. Krasnova; Subramaniam Jayanthi; Johnalyn Lyles

The amphetamines, including amphetamine (AMPH), methamphetamine (METH) and 3,4-methylenedioxymethamphetamine (MDMA), are among abused drugs in the US and throughout the world. Their abuse is associated with severe neurologic and psychiatric adverse events including the development of psychotic states. These neuropsychiatric complications might, in part, be related to drug-induced neurotoxic effects, which include damage to dopaminergic and serotonergic terminals, neuronal apoptosis, as well as activated astroglial and microglial cells in the brain. The purpose of the present review is to summarize the toxic effects of AMPH, METH and MDMA. The paper also presents some of the factors that are thought to underlie this toxicity. These include oxidative stress, hyperthermia, excitotoxicity and various apoptotic pathways. Better understanding of the cellular and molecular mechanisms involved in their toxicity should help to generate modern therapeutic approaches to prevent or attenuate the long-term consequences of amphetamine use disorders in humans.


Biological Psychiatry | 2010

Prenatal Interaction of Mutant DISC1 and Immune Activation Produces Adult Psychopathology

Bagrat Abazyan; Jun Nomura; Geetha Kannan; Koko Ishizuka; Kellie L. Tamashiro; Frederick Nucifora; Vladimir Pogorelov; Bruce Ladenheim; Chunxia Yang; Irina N. Krasnova; Jean Lud Cadet; Carlos Pardo; Susumu Mori; Atsushi Kamiya; Michael W. Vogel; Akira Sawa; Christopher A. Ross; Mikhail V. Pletnikov

BACKGROUND Gene-environment interactions (GEI) are involved in the pathogenesis of mental diseases. We evaluated interaction between mutant human disrupted-in-schizophrenia 1 (mhDISC1) and maternal immune activation implicated in schizophrenia and mood disorders. METHODS Pregnant mice were treated with saline or polyinosinic:polycytidylic acid at gestation day 9. Levels of inflammatory cytokines were measured in fetal and adult brains; expression of mhDISC1, endogenous DISC1, lissencephaly type 1, nuclear distribution protein nudE-like 1, glycoprotein 130, growth factor receptor-bound protein 2, and glycogen synthase kinase-3beta were assessed in cortical samples of newborn mice. Tissue content of monoamines, volumetric brain abnormalities, dendritic spine density in the hippocampus, and various domains of the mouse behavior repertoire were evaluated in adult male mice. RESULTS Prenatal interaction produced anxiety, depression-like responses, and altered social behavior that were accompanied by decreased reactivity of the hypothalamic-pituitary-adrenal axis, attenuated serotonin neurotransmission in the hippocampus, reduced enlargement of lateral ventricles, decreased volumes of amygdala and periaqueductal gray matter and density of spines on dendrites of granule cells of the hippocampus. Prenatal interaction modulated secretion of inflammatory cytokines in fetal brains, levels of mhDISC1, endogenous mouse DISC1, and glycogen synthase kinase-3beta. The behavioral effects of GEI were observed only if mhDISC1 was expressed throughout the life span. CONCLUSIONS Prenatal immune activation interacted with mhDISC1 to produce the neurobehavioral phenotypes that were not seen in untreated mhDISC1 mice and that resemble aspects of major mental illnesses. Our DISC1 mouse model is a valuable system to study GEI relevant to mental illnesses.


International Review of Neurobiology | 2009

Molecular Bases of Methamphetamine-Induced Neurodegeneration

Jean Lud Cadet; Irina N. Krasnova

Methamphetamine (METH) is a highly addictive psychostimulant drug, whose abuse has reached epidemic proportions worldwide. The addiction to METH is a major public concern because its chronic abuse is associated with serious health complications including deficits in attention, memory, and executive functions in humans. These neuropsychiatric complications might, in part, be related to drug-induced neurotoxic effects, which include damage to dopaminergic and serotonergic terminals, neuronal apoptosis, as well as activated astroglial and microglial cells in the brain. Thus, the purpose of the present paper is to review cellular and molecular mechanisms that might be responsible for METH neurotoxicity. These include oxidative stress, activation of transcription factors, DNA damage, excitotoxicity, blood-brain barrier breakdown, microglial activation, and various apoptotic pathways. Several approaches that allow protection against METH-induced neurotoxic effects are also discussed. Better understanding of the cellular and molecular mechanisms involved in METH toxicity should help to generate modern therapeutic approaches to prevent or attenuate the long-term consequences of psychostimulant use disorders in humans.


PLOS ONE | 2010

Methamphetamine Self-Administration Is Associated with Persistent Biochemical Alterations in Striatal and Cortical Dopaminergic Terminals in the Rat

Irina N. Krasnova; Zuzana Justinova; Bruce Ladenheim; Subramaniam Jayanthi; Michael T. McCoy; Chanel Barnes; John E. Warner; Steven R. Goldberg; Jean Lud Cadet

Methamphetamine (meth) is an illicit psychostimulant that is abused throughout the world. Repeated passive injections of the drug given in a single day or over a few days cause significant and long-term depletion of dopamine and serotonin in the mammalian brain. Because meth self-administration may better mimic some aspects of human drug-taking behaviors, we examined to what extent this pattern of drug treatment might also result in damage to monoaminergic systems in the brain. Rats were allowed to intravenously self-administer meth (yoked control rats received vehicle) 15 hours per day for 8 days before being euthanized at either 24 hours or at 7 and 14 days after cessation of drug taking. Meth self-administration by the rats was associated with a progressive escalation of daily drug intake to 14 mg/kg per day. Animals that self-administered meth exhibited dose-dependent decreases in striatal dopamine levels during the period of observation. In addition, there were significant reductions in the levels of striatal dopamine transporter and tyrosine hydroxylase proteins. There were also significant decreases in the levels of dopamine, dopamine transporter, and tyrosine hydroxylase in the cortex. In contrast, meth self-administration caused only transient decreases in norepinephrine and serotonin levels in the two brain regions, with these values returning to normal at seven days after cessation of drug taking. Importantly, meth self-administration was associated with significant dose-dependent increases in glial fibrillary acidic protein in both striatum and cortex, with these changes being of greater magnitude in the striatum. These results suggest that meth self-administration by rats is associated with long-term biochemical changes that are reminiscent of those observed in post-mortem brain tissues of chronic meth abusers.


Biological Psychiatry | 2014

Methamphetamine downregulates striatal glutamate receptors via diverse epigenetic mechanisms.

Subramaniam Jayanthi; Michael T. McCoy; Billy T. Chen; Jonathan P. Britt; Saïd Kourrich; Hau-Jie Yau; Bruce Ladenheim; Irina N. Krasnova; Antonello Bonci; Jean Lud Cadet

BACKGROUND Chronic methamphetamine (METH) exposure causes neuroadaptations at glutamatergic synapses. METHODS To identify the METH-induced epigenetic underpinnings of these neuroadaptations, we injected increasing METH doses to rats for 2 weeks and measured striatal glutamate receptor expression. We then quantified the effects of METH exposure on histone acetylation. We also measured METH-induced changes in DNA methylation and DNA hydroxymethylation. RESULTS Chronic METH decreased transcript and protein expression of GluA1 and GluA2 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) and GluN1 N-methyl-D-aspartate receptor subunits. These changes were associated with altered electrophysiological glutamatergic responses in striatal neurons. Chromatin immunoprecipitation-polymerase chain reaction revealed that METH decreased enrichment of acetylated histone H4 on GluA1, GluA2, and GluN1 promoters. Methamphetamine exposure also increased repressor element-1 silencing transcription factor (REST) corepressor 1, methylated CpG binding protein 2, and histone deacetylase 2 enrichment, but not of sirtuin 1 or sirtuin 2, onto GluA1 and GluA2 gene sequences. Moreover, METH caused interactions of REST corepressor 1 and methylated CpG binding protein 2 with histone deacetylase 2 and of REST with histone deacetylase 1. Surprisingly, methylated DNA immunoprecipitation and hydroxymethylated DNA immunoprecipitation-polymerase chain reaction revealed METH-induced decreased enrichment of 5-methylcytosine and 5-hydroxymethylcytosine at GluA1 and GluA2 promoter sequences. Importantly, the histone deacetylase inhibitor, valproic acid, blocked METH-induced decreased expression of AMPAR and N-methyl-D-aspartate receptor subunits. Finally, valproic acid also attenuated METH-induced decrease H4K16Ac recruitment on AMPAR gene sequences. CONCLUSIONS These observations suggest that histone H4 hypoacetylation may be the main determinant of METH-induced decreased striatal glutamate receptor expression.


Neurobiology of Disease | 2013

CREB phosphorylation regulates striatal transcriptional responses in the self-administration model of methamphetamine addiction in the rat

Irina N. Krasnova; Margarit Chiflikyan; Zuzana Justinova; Michael T. McCoy; Bruce Ladenheim; Subramaniam Jayanthi; Cynthia Quintero; Christie Brannock; Chanel Barnes; Jordan E. Adair; Elin Lehrmann; Firas Kobeissy; Mark S. Gold; Kevin G. Becker; Steven R. Goldberg; Jean Lud Cadet

Neuroplastic changes in the dorsal striatum participate in the transition from casual to habitual drug use and might play a critical role in the development of methamphetamine (METH) addiction. We examined the influence of METH self-administration on gene and protein expression that may form substrates for METH-induced neuronal plasticity in the dorsal striatum. Male Sprague-Dawley rats self-administered METH (0.1mg/kg/injection, i.v.) or received yoked saline infusions during eight 15-h sessions and were euthanized 2h, 24h, or 1month after cessation of METH exposure. Changes in gene and protein expression were assessed using microarray analysis, RT-PCR and Western blots. Chromatin immunoprecipitation (ChIP) followed by PCR was used to examine epigenetic regulation of METH-induced transcription. METH self-administration caused increases in mRNA expression of the transcription factors, c-fos and fosb, the neurotrophic factor, Bdnf, and the synaptic protein, synaptophysin (Syp) in the dorsal striatum. METH also caused changes in ΔFosB, BDNF and TrkB protein levels, with increases after 2 and 24h, but decreases after 1month of drug abstinence. Importantly, ChIP-PCR showed that METH self-administration caused enrichment of phosphorylated CREB (pCREB), but not of histone H3 trimethylated at lysine 4 (H3K4me3), on promoters of c-fos, fosb, Bdnf and Syp at 2h after cessation of drug intake. These findings show that METH-induced changes in gene expression are mediated, in part, by pCREB-dependent epigenetic phenomena. Thus, METH self-administration might trigger epigenetic changes that mediate alterations in expression of genes and proteins serving as substrates for addiction-related synaptic plasticity.


The FASEB Journal | 2005

Amphetamine induces apoptosis of medium spiny striatal projection neurons via the mitochondria-dependent pathway

Irina N. Krasnova; Bruce Ladenheim; Jean Lud Cadet

Amphetamine (AMPH) is a psychostimulant whose chronic abuse may cause impairments in attention and memory in humans. These cognitive deficits might be related to neurotoxic effects of the drug. One such toxic effect is the well‐described destruction of striatal dopaminergic terminals in mammals. In the present study, we investigated the possibility that AMPH might also cause neuronal apoptosis in the rodent striatum. Administration of a dose of the drug (10 mg/kg, 4 times, every 2 h) that is toxic to dopaminergic terminals resulted in the appearance of striatal cells that were positive for cleaved caspase‐3 and for terminal deoxynucleotidyl transferase‐mediated biotin‐dUTP nick‐end labeling (TUNEL), observations that are indicative of an ongoing apoptotic process. Dual immunofluorescence staining revealed that cleaved caspase‐3‐positive cells express calbindin and DARPP‐32, but not somatostatin, parvalbumin, or cholinergic markers. In addition, AMPH also caused increased expression of p53 and Bax at both transcript and protein levels; in contrast, Bcl‐2 levels were decreased after the AMPH injections. Moreover, Bax knockout mice showed resistance to AMPH‐induced apoptotic cell death but not to AMPH‐induced destruction of dopaminergic terminals. When taken together, these observations indicate that injections of doses of AMPH that are known to destroy striatal dopamine terminals can also cause apoptotic death of postsynaptic medium spiny projection neurons via mitochondria‐dependent mechanisms.


Biological Psychiatry | 2009

Methamphetamine- and Trauma-Induced Brain Injuries: Comparative Cellular and Molecular Neurobiological Substrates

Mark S. Gold; Firas Kobeissy; Kevin K. W. Wang; Lisa J. Merlo; Adriaan W. Bruijnzeel; Irina N. Krasnova; Jean Lud Cadet

The use of methamphetamine (METH) is a growing public health problem, because its abuse is associated with long-term biochemical and structural effects on the human brain. Neurodegeneration is often observed in humans, because of mechanical injuries (e.g., traumatic brain injury [TBI]) and ischemic damage (strokes). In this review, we discuss recent findings documenting the fact that the psychostimulant drug METH can cause neuronal damage in several brain regions. The accumulated evidence from our laboratories and those of other investigators indicates that acute administration of METH leads to activation of calpain and caspase proteolytic systems. These systems are also involved in causing neuronal damage secondary to traumatic and ischemic brain injuries. Protease activation is accompanied by proteolysis of endogenous neuronal structural proteins (alphaII-spectrin protein and microtubule-associated protein-tau), evidenced by the appearance of their breakdown products after these injuries. When taken together, these observations suggest that METH exposure, like TBI, can cause substantial damage to the brain by causing both apoptotic and necrotic cell death in the brains of METH addicts who use large doses of the drug during their lifetimes. Finally, because METH abuse is accompanied by functional and structural changes in the brain similar to those in TBI, METH addicts might experience greater benefit if their treatment involved greater emphasis on rehabilitation in conjunction with potential neuroprotective pharmacological agents such as calpain and caspase inhibitors similar to those used in TBI.


Neuroscience | 2012

Sex-specific changes in gene expression and behavior induced by chronic Toxoplasma infection in mice.

Jianchun Xiao; Geetha Kannan; Lorraine Jones-Brando; C. Brannock; Irina N. Krasnova; Jean-Lud Cadet; Mikhail V. Pletnikov; Robert H. Yolken

There is growing evidence that Toxoplasma gondii modifies the behavior of its intermediate hosts. We investigated the molecular basis of these infection-induced behavioral changes, followed by five related behavioral tests to assess the extent of biological relevance. Gene expression signatures were generated in the frontal cortex of male and female mice during the latent stage of infection. We found marked sex-dependent expression differences in mice. In female mice, Toxoplasma infection altered the expression of genes involved in the development of the forebrain, neurogenesis, and sensory and motor coordination (i.e. downregulation of fatty acid-binding protein 7 and eyes absent homolog 1, upregulation of semaphorin 7A). In male mice, infection led mainly to modulation of genes associated with olfactory function (i.e. downregulation of a number of olfactory receptors and dopamine receptor D4, upregulation of slit homolog 1). Although infection appears to affect the olfactory function in male mice, it is the female but not male mice that exhibited attraction to cat odor. In contrast, infected male mice showed a deficit in social transmission of food preference. In contrast to males, infected females displayed locomotor hyperactivity in open field. General olfaction and sensorimotor gating were normal in both male and female infection. Our results indicate that the sex of the host plays a major role in determining variable brain and behavior changes following Toxoplasma infection. These observations are consistent with heterogeneity of neuropsychiatric outcomes of the infection in humans.

Collaboration


Dive into the Irina N. Krasnova's collaboration.

Top Co-Authors

Avatar

Jean Lud Cadet

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Bruce Ladenheim

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Michael T. McCoy

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Subramaniam Jayanthi

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Christie Brannock

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin G. Becker

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Genevieve Beauvais

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Mikhail V. Pletnikov

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Elin Lehrmann

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge