Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 107: 58-67, 2016 08.
Article in English | MEDLINE | ID: mdl-26921770

ABSTRACT

There has been increasing interest in the rostromedial tegmental nucleus (RMTg), given its potential regulatory role in many aversion-related behaviors. The RMTg contains mostly GABAergic neurons, sends a dense inhibitory projection to dopamine neurons in the midbrain, and is rich with µ-opioid receptors (MOR). Like most addictive drugs, ethanol has both aversive and rewarding properties. However, the cellular mechanisms underlying the effects of ethanol, particularly the aversive effect that limits its intake are not well understood. Recent studies have linked aversion with synaptic inhibition of dopamine neurons in the ventral tegmental area. To determine a potential role that the RMTg plays in the effect of ethanol, in this study, we employed a neurotoxin, dermorphin-saporin (DS), to lesion RMTg neurons prior to assessing ethanol-related behaviors. Rats were infused with DS bilaterally into the RMTg. This manipulation substantially increased the intake and preference for ethanol but not sucrose. It also reduced the number of neurons with MOR and glutamic acid decarboxylase 67 immunoreactivity within the RMTg. These changes did not occur after intra-RMTg infusion of blank saporin or vehicle. Importantly, intra-RMTg DS infusion significantly enhanced expression of conditioned place preference induced by ethanol (2 g/kg, i.p.), and slowed the extinction process. These results suggest that MOR-expressing GABAergic neurons in the RMTg contribute significantly to the regulation of ethanol consumption and related behaviors.


Subject(s)
Alcohol Drinking/metabolism , Alcohol-Related Disorders/metabolism , GABAergic Neurons/metabolism , Receptors, Opioid, mu/metabolism , Tegmentum Mesencephali/metabolism , Alcohol Drinking/pathology , Alcohol-Related Disorders/pathology , Animals , Central Nervous System Depressants/administration & dosage , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Disease Models, Animal , Ethanol/administration & dosage , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Female , GABAergic Neurons/drug effects , GABAergic Neurons/pathology , Glutamate Decarboxylase/metabolism , Motor Activity/drug effects , Motor Activity/physiology , Opioid Peptides , Random Allocation , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1 , Saporins , Self Administration , Spatial Behavior/drug effects , Spatial Behavior/physiology , Tegmentum Mesencephali/drug effects , Tegmentum Mesencephali/pathology
2.
Brain Sci ; 4(2): 356-75, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24961766

ABSTRACT

Propofol is a general anesthetic widely used in surgical procedures, including those in pregnant women. Preclinical studies suggest that propofol may cause neuronal injury to the offspring of primates if it is administered during pregnancy. However, it is unknown whether those neuronal changes would lead to long-term behavioral deficits in the offspring. In this study, propofol (0.4 mg/kg/min, IV, 2 h), saline, or intralipid solution was administered to pregnant rats on gestational day 18. We detected increased levels of cleaved caspase-3 in fetal brain at 6 h after propofol exposure. The neuronal density of the hippocampus of offspring was reduced significantly on postnatal day 10 (P10) and P28. Synaptophysin levels were also significantly reduced on P28. Furthermore, exploratory and learning behaviors of offspring rats (started at P28) were assessed in open-field trial and eight-arm radial maze. The offspring from propofol-treated dams showed significantly less exploratory activity in the open-field test and less spatial learning in the eight-arm radial maze. Thus, this study suggested that propofol exposure during pregnancy in rat increased cleaved caspsase-3 levels in fetal brain, deletion of neurons, reduced synaptophysin levels in the hippocampal region, and persistent learning deficits in the offspring.

3.
Neurotoxicol Teratol ; 43: 51-8, 2014.
Article in English | MEDLINE | ID: mdl-24726880

ABSTRACT

Preclinical studies suggest that propofol may cause damage to immature neurons. However, the effect of maternal propofol exposure on the neuronal development of the offspring is largely unknown. In this study, pregnant rats were assigned to receive continuous infusion of saline (control) or propofol for 1 h (1HP) or 2 h (2HP) on gestational day 18. An additional group (lipid) was assigned to receive continuous infusion of intralipid fat emulsion (vehicle of propofol) for 2 h. Pups were then tested on the appearance and progression of sensory and physical motor abilities between postnatal day 1 (P1) and P28. The brain and body weights of pups from 2HP group on P10 were significantly lower than those from the saline control group, although they were the same in all four groups at birth (P0). Pups from 1HP and 2HP groups, but not lipid group, showed slower maturation of eyes (delayed opening) and several neurological reflexes (hindlimb reflex, righting reflex); they also showed delayed improvement in execution on gait reflex and inclined board tests. The forelimb reflex and negative geotaxis were also delayed in 2HP group. All parameters examined except body weight of 2HP pups recovered to normal levels by P28. We conclude that administration of propofol to pregnant rats leads to retardation in physical and neurological reflex development in their offspring.


Subject(s)
Developmental Disabilities/etiology , Hypnotics and Sedatives/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Propofol/toxicity , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Blood Gas Analysis , Disease Models, Animal , Female , Male , Maternal Behavior , Pregnancy , Rats , Rats, Sprague-Dawley , Reflex/physiology , Time Factors
4.
PLoS One ; 8(5): e64473, 2013.
Article in English | MEDLINE | ID: mdl-23717619

ABSTRACT

Interleukin (IL)-2, a T-helper 1 (Th1) cell-derived cytokine, which potently modulates dopamine activity and neuronal excitability in mesolimbic structures, is linked with pathological outcomes (e.g., schizophrenia, depression, etc.) that at least partly reflect alterations in central dopaminergic processes. It has been suggested that dopamine neurons undergo pruning during adolescence and abnormalities in pruning predispose individuals to behavioral disorders. Since IL-2 is known as a neurodevelopmental factor affecting associated behavioral processes, the present study tested whether IL-2 can modulate stereotypic behaviors in both the periadolescent and adult periods. This study determined whether IL-2 treatment would produce long-lasting changes in sensitivity to a later challenge with IL-2 or GBR 12909, a highly selective dopamine uptake inhibitor. Four experiments were conducted. Firstly, a decrease in novelty-induced stereotypic behavior was observed in BALB/c periadolescent mice (38 days of age) following IL-2 administration (0.4 µg/2 ml) relative to vehicle control. In the second experiment, an initial dose of IL-2 was given in the periadolescent period, but did not affect rearing responses. A second dose of IL-2 given to the animals 30 days later as adults, resulted in a significant increase in rearing behaviors relative to control animals. In the third experiment, separate groups of experimental and control mice were administered GBR 12909, a highly selective dopamine reuptake inhibitor, 30 days following treatment with either IL-2 or vehicle. It was noted that this experimental group, which initially received IL-2, exhibited stereotypy, as evidenced by increased sniffing behavior. A fourth experiment revealed that IL-2 administered in periadolesecence and adulthood had no effect on other motor responses, indicating that IL-2 selectively modulates selective stereotypic behaviors. The results provide evidence, for the first time, that long-term changes in stereotypy in periadolescent mice are linked to an IL-2 mechanism, possibly utilizing dopamine.


Subject(s)
Dopamine Uptake Inhibitors/metabolism , Dopamine Uptake Inhibitors/pharmacology , Interleukin-2/metabolism , Interleukin-2/pharmacology , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Dopamine Uptake Inhibitors/administration & dosage , Female , Interleukin-2/administration & dosage , Male , Mice , Motor Activity/drug effects , Time Factors
5.
PLoS One ; 7(7): e41623, 2012.
Article in English | MEDLINE | ID: mdl-22911828

ABSTRACT

Soluble cytokine receptors are normal constituents of body fluids that regulate peripheral cytokine and lymphoid activity and whose levels are increased in states of immune activation. Soluble interleukin-6 receptor (sIL-6R) levels positively correlate with disease progression in some autoimmune conditions and psychiatric disorders. Particularly strong links between levels of sIL-6R and the severity of psychotic symptoms occur in schizophrenia, raising the possibility that sIL-6R is involved in this disease. However, there is no evidence that peripheral sIL-6R induces relevant behavioral disturbances. We showed that single subcutaneous injections of sIL-6R (0-1 µg), stimulated novelty stress-induced exploratory motor behaviors in male Balb/c mice within 20-40-min of injection. A progressive increase in vertical stereotypies was observed 40-80 min post injection, persisting for the remainder of the test session. Paralleling these stimulant-like effects, sIL-6R pre-treatment significantly enhanced stereotypy scores following challenge with GBR 12909. We found that peripherally administered sIL-6R crossed the blood-brain barrier, localizing in brain regions associated with cortico-striatal-thalamo-cortical (CSTC) circuits, which are putative neuroanatomical substrates of disorders associated with repetitive stereotypies. Peripherally administered sIL-6R co-localized with gp130, a transmembrane protein involved in IL-6 trans-signaling, in the nucleus accumbens, caudate-putamen, motor and infralimbic cortices, and thalamic nuclei, but not with gp130 in the ventral tegmental area, substantia nigra, or sensorimotor cortex,. The results suggest that peripheral sIL-6R can act as a neuroimmune messenger, crossing the blood brain barrier (BBB) to selectively target CSTC circuits rich in IL-6 trans-signaling protein, and inducing repetitive stereotypies. As such sIL-6R may represent a novel therapeutic agent for relevant psychiatric disorders.


Subject(s)
Cerebral Cortex/metabolism , Cytokine Receptor gp130/metabolism , Motor Activity , Neostriatum/metabolism , Nerve Net/metabolism , Receptors, Interleukin-6/metabolism , Thalamus/metabolism , Animals , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Humans , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Neostriatum/drug effects , Nerve Net/drug effects , Piperazines/pharmacology , Protein Transport/drug effects , Receptors, Interleukin-6/administration & dosage , Solubility/drug effects , Staining and Labeling , Thalamus/drug effects
6.
Differentiation ; 84(2): 214-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22824626

ABSTRACT

Mesenchymal stromal cells (MSCs) show promise for treatment of a variety of neurological and other disorders. Cat has a high degree of linkage with the human genome and has been used as a model for analysis of neurological disorders such as stroke, Alzheimer's disease and motor disorders. The present study was designed to characterize bone marrow-derived MSCs from cats and to investigate the capacity to generate functional peptidergic neurons. MSCs were expanded with cells from the femurs of cats and then characterized by phenotype and function. Phenotypically, feline and human MSCs shared surface markers, and lacked hematopoietic markers, with similar morphology. As compared to a subset of human MSCs, feline MSCs showed no evidence of the major histocompatibility class II. Since the literature suggested Stro-1 as an indicator of pluripotency, we compared early and late passages feline MSCs and found its expression in >90% of the cells. However, the early passage cells showed two distinct populations of Stro-1-expressing cells. At passage 5, the MSCs were more homogeneous with regards to Stro-1 expression. The passage 5 MSCs differentiated to osteogenic and adipogenic cells, and generated neurons with electrophysiological properties. This correlated with the expression of mature neuronal markers with concomitant decrease in stem cell-associated genes. At day 12 induction, the cells were positive for MAP2, Neuronal Nuclei, tubulin ßIII, Tau and synaptophysin. This correlated with electrophysiological maturity as presented by excitatory postsynaptic potentials (EPSPs). The findings indicate that the cat may constitute a promising biomedical model for evaluation of novel therapies such as stem cell therapy in such neurological disorders as Alzheimer's disease and stroke.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Neurogenesis , Neurons/metabolism , Phenotype , Adipogenesis , Animals , Antigens, Surface/metabolism , Cats , Cells, Cultured , Excitatory Postsynaptic Potentials , Humans , Neurons/cytology , Osteogenesis , Species Specificity
7.
PLoS One ; 7(4): e36316, 2012.
Article in English | MEDLINE | ID: mdl-22558434

ABSTRACT

Soluble cytokine receptors are normal constituents of body fluids that regulate peripheral cytokine and lymphoid activity. Levels of soluble IL-2 receptors (sIL-2R) are elevated in psychiatric disorders linked with autoimmune processes, including ones in which repetitive stereotypic behaviors and motor disturbances are present. However, there is no evidence that sIL-2Rs (or any peripheral soluble receptor) induce such behavioral changes, or that they localize in relevant brain regions. Here, we determined in male Balb/c mice the effects of single peripheral injections of sIL-2Rα or sIL-2Rß (0-2 µg/male Balb/c mouse; s.c.) on novelty-induced ambulatory activity and stereotypic motor behaviors. We discovered that sIL-2Rα increased the incidence of in-place stereotypic motor behaviors, including head up head bobbing, rearing/sniffing, turning, and grooming behavior. A wider spectrum of behavioral changes was evident in sIL-2Rß-treated mice, including increases in vertical and horizontal ambulatory activity and stereotypic motor movements. To our knowledge, this is the first demonstration that soluble receptors induce such behavioral disturbances. In contrast, soluble IL-1 Type-1 receptors (0-4 µg, s.c.) didn't appreciably affect these behaviors. We further demonstrated that sIL-2Rα and sIL-2Rß induced marked increases in c-Fos in caudate-putamen, nucleus accumbens and prefrontal cortex. Anatomical specificity was supported by the presence of increased activity in lateral caudate in sIL-2Rα treated mice, while sIL-2Rß treated mice induced greater c-Fos activity in prepyriform cortex. Moreover, injected sIL-2Rs were widely distributed in regions that showed increased c-Fos expression. Thus, sIL-2Rα and sIL-2Rß induce marked subunit- and soluble cytokine receptor-specific behavioral disturbances, which included increases in the expression of ambulatory activity and stereotypic motor behaviors, while inducing increased neuronal activity localized to cortex and striatum. These findings suggest that sIL-2Rs act as novel immune-to- brain messengers and raise the possibility that they contribute to the disease process in psychiatric disorders in which marked increases in these receptors have been reported.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Interleukin-2 Receptor beta Subunit/pharmacology , Animals , Behavior, Animal/drug effects , Humans , Interleukin-2 Receptor alpha Subunit/chemistry , Interleukin-2 Receptor beta Subunit/chemistry , Male , Mice , Mice, Inbred BALB C , Neostriatum/drug effects , Neostriatum/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Solubility
8.
J Neuroimmunol ; 246(1-2): 58-64, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22464399

ABSTRACT

Guillain-Barré syndrome (GBS) is an autoimmune-mediated disease triggered by a preceding infection. A substantial body of evidence implicates antibodies to various gangliosides in subtypes of GBS. A significant proportion of patients with acute demyelinating subset of GBS have IgG antibodies against peripheral nervous system myelin specific neolactogangliosides such as LM1 and Hex-LM1. Although anti-neolactoganglioside antibodies in GBS were described more than two decades ago, their pathogenic role in neuropathy remains unknown due to the lack of suitable experimental models. In this study, we immunized ten guinea pigs with purified LM1 ganglioside mixed with keyhole limpet hemocyanin (KLH) and emulsified in complete Freund's adjuvant (CFA). Control guinea pigs were injected with KLH emulsified in CFA only. The animals were bled every four week intervals. The animals were boosted 3 times every four weeks. Experiments were terminated four months after initial immunization. Nine of 10 guinea pigs immunized with LM1 exhibited antibody responses to LM1. Anti-LM1 IgG titers in nine guinea pigs ranged from 1:400 to 1:12,800 at 16-weeks after initial immunization. Anti-LM1 antibodies were predominantly of IgG2 subclass. One guinea pig with the highest levels of IgG antibodies exhibited mild signs of neuropathy. There was no evidence of demyelination or inflammation in the sciatic nerves of LM1-immunized guinea pigs. Anti-LM1 antibodies bound to rat sciatic nerve myelin and to isolated rat Schwann cells. In summary, our findings suggest that relatively high levels of anti-LM1 IgG antibodies can be induced in guinea pigs and that LM1 is localized in peripheral nerve myelin and in Schwann cells. Further studies are needed to determine the pathogenic potential of anti-neolactoganglioside antibodies in neuropathy.


Subject(s)
Autoantibodies/biosynthesis , Gangliosides/immunology , Immunoglobulin G/biosynthesis , Animals , Autoantibodies/metabolism , Demyelinating Diseases/diagnosis , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Freund's Adjuvant/administration & dosage , Gangliosides/administration & dosage , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/pathology , Guinea Pigs , Hemocyanins/administration & dosage , Hemocyanins/immunology , Humans , Immunization/methods , Immunoglobulin G/metabolism , Schwann Cells/immunology , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/immunology , Sciatic Nerve/pathology
9.
Brain Behav Immun ; 26(4): 521-33, 2012 May.
Article in English | MEDLINE | ID: mdl-22285613

ABSTRACT

Group A beta-hemolytic streptococcus (GABHS) infections are implicated in neuropsychiatric disorders associated with an increased expression of repetitive stereotyped movements. Anti-streptococcus IgG presumably cross-reacts with elements on basal ganglia cells, modifies their function, and triggers symptoms. IgM may play a unique role in precipitating behavioral disturbances since variations in cortico-striatal activity occur in temporal congruity with peak IgM titers during an orchestrated immune response. We discovered in Balb/c mice that single subcutaneous injections of mouse monoclonal IgM antibodies to streptococcus group A bacteria induce marked dose-dependent increases in repetitive stereotyped movements, including head bobbing, sniffing, and intense grooming. Effects were antibody- and antigen-specific: anti-streptococcus IgG stimulated ambulatory activity and vertical activity but not these stereotypies, while anti-KLH IgM reduced activity. We suggest that anti-streptococcus IgM and IgG play unique roles in provoking GABHS-related behavioral disturbances. Paralleling its stereotypy-inducing effects, anti-streptococcus IgM stimulated Fos-like immunoreactivity in regions linked to cortico-striatal projections involved in motor control, including subregions of the caudate, nucleus accumbens, and motor cortex. This is the first evidence that anti-streptococcus IgM antibodies induce in vivo functional changes in these structures. Moreover, there was a striking similarity in the distributions of anti-streptococcus IgM deposits and Fos-like immunoreactivity in these regions. Of further importance, Fcα/µ receptors, which bind IgM, were present- and co-localized with anti-streptococcus IgM in these structures. We suggest that anti-streptococcus IgM-induced alterations of cell activity reflect local actions of IgM that involve Fcα/µ receptors. These findings support the use of anti-streptococcus monoclonal antibody administration in Balb/c mice to model GABHS-related behavioral disturbances and identify underlying mechanisms.


Subject(s)
Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunoglobulin M/pharmacology , Stereotyped Behavior/drug effects , Stereotypic Movement Disorder/immunology , Streptococcus pyogenes/immunology , Animals , Basal Ganglia/drug effects , Basal Ganglia/immunology , Hemocyanins/immunology , Immunoglobulin G/pharmacology , Immunoglobulin M/immunology , Male , Mice , Mice, Inbred BALB C , Models, Animal , Motor Cortex/drug effects , Motor Cortex/immunology , Proto-Oncogene Proteins c-fos/immunology , Receptors, Fc/immunology
10.
Int J Law Psychiatry ; 34(1): 20-9, 2011.
Article in English | MEDLINE | ID: mdl-21112635

ABSTRACT

The present paper addresses the philosophical problem raised by current causal neurochemical models of impulsive violence and aggression: to what extent can we hold violent criminal offenders responsible for their conduct if that conduct is the result of deterministic biochemical processes in the brain. This question is currently receiving a great deal of attention among neuroscientists, legal scholars and philosophers. We examine our current knowledge of neuroscience to assess the possible roles of deterministic factors which induce impulsive aggression, and the extent to which this behavior can be controlled by neural conditioning mechanisms. Neural conditioning mechanisms, we suggest, may underlie what we consider the basis of responsible (though not necessarily moral) behavior: the capacity to give and take reasons. The models we first examine are based in part upon the role played by the neurotransmitter, serotonin, in the regulation of violence and aggression. Collectively, these results would appear to argue in favor of the view that low brain serotonin levels induce impulsive aggression which overrides mechanisms related to rational decision making processes. We next present an account of responsibility as based on the capacity to exercise a certain kind of reason-responsive control over one's conduct. The problem with such accounts of responsibility, however, is that they fail to specify a neurobiological realization of such mechanisms of control. We present a neurobiological, and weakly determinist, framework for understanding how persons can exercise guidance control over their conduct. This framework is based upon classical conditioning of neurons in the prefrontal cortex that allow for a decision making mechanism that provides for prefrontal cortical control of the sites in the brain which express aggressive behavior that include the hypothalamus and midbrain periaqueductal gray. The authors support the view that, in many circumstances, neural conditioning mechanisms provide the basis for the control of human aggression in spite of the presence of brain serotonin levels that might otherwise favor the expression of impulsive aggressive behavior. Indeed if those neural conditioning mechanisms underlie the human capacity to exercise control, they may be the neural realization of reason-responsiveness generally.


Subject(s)
Aggression/physiology , Personal Autonomy , Serotonin Receptor Agonists/blood , Serotonin/blood , Brain/metabolism , Female , Hostility , Humans , Impulsive Behavior/chemically induced , Male , Serotonin/pharmacology , Serotonin Receptor Agonists/pharmacology
11.
Brain Behav Immun ; 24(8): 1276-80, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20685290

ABSTRACT

The mechanisms underlying violence and aggression and its control remain poorly understood. Using the resident-intruder paradigm, we have discovered that resident mice with combined deletion of TNF receptor type 1 (TNF-R1) and type 2 (TNF-R2) genes show a striking absence of aggressive behavior, which includes fighting, sideways postures, and tail rattling. In parallel, resident TNF-R1 and TNF-R2 knockout mice show an increase in non-aggressive exploration of the intruder mice. Given the relationship between aggression and anxiety, we also measured anxiety-related behavior, as reflected by performance in the Open Field and the Light-Dark Choice Test. Compared with wild type mice, TNF-R1 and TNF-R2 deficient mice spent significantly more time and showed increased movement in the center of the Open Field and in the illuminated compartment of the light-dark box, suggesting an anxiolytic-like profile. Together, these data show that combined deletion of TNF-R1 and TNF-R2 results in a striking absence of aggressive behavior, an increase in non-aggressive exploration, and anxiolytic-like effects. These findings identify potent roles for TNF in regulating aggression and anxiety-related behavior, and suggest that TNF receptor signaling tonically modulates activity in brain regions underlying these behaviors.


Subject(s)
Aggression/physiology , Anxiety/psychology , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Animals , Choice Behavior , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Receptors, Tumor Necrosis Factor, Type II/physiology
12.
Int J Law Psychiatry ; 32(4): 209-15, 2009.
Article in English | MEDLINE | ID: mdl-19596153

ABSTRACT

The present paper reviews and summarizes the basic findings concerning the nature of the neurobiological and behavioral characteristics of aggression and rage. For heuristic purposes, the types of aggression will be reduced to two categories - defensive rage (affective defense) and predatory attack. This approach helps explain both the behavioral properties of aggression as well as the underlying neural substrates and mechanisms of aggression both in animals and humans. Defensive rage behavior is activated by a threatening stimulus that is real or perceived and is associated with marked sympathetic output. This yields impulsivity with minimal cortical involvement. Predatory attack behavior in both animals and humans is generally planned, taking minutes, hours, days, weeks, months, or even years (with respect to humans) for it to occur and is directed upon a specific individual target; it reflects few outward sympathetic signs and is believed to require cortical involvement for its expression. Predatory attack requires activation of the lateral hypothalamus, while defensive rage requires activation of the medial hypothalamus and midbrain periaqueductal gray (PAG). Both forms of aggressive behavior are controlled by components of the limbic system, a region of the forebrain that is influenced by sensory inputs from the cerebral cortex and monoaminergic inputs from the brainstem reticular formation. Control of aggressive tendencies is partly modifiable through conditioning and related learning principles generated through the cerebral cortex.


Subject(s)
Aggression/psychology , Neurosciences , Aggression/physiology , Animals , Biogenic Amines , Defense Mechanisms , Humans , Limbic System , Predatory Behavior/physiology , Serotonin , Violence
13.
Brain Behav Immun ; 22(2): 224-33, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17890051

ABSTRACT

Feline defensive rage, a form of aggressive behavior that occurs in response to a threat can be elicited by electrical stimulation of the medial hypothalamus or midbrain periaqueductal gray (PAG). Our laboratory has recently begun a systematic examination of the role of cytokines in the regulation of rage and aggressive behavior. It was shown that the cytokine, interleukin-2 (IL-2), differentially modulates defensive rage when microinjected into the medial hypothalamus and PAG by acting through separate neurotransmitter systems. The present study sought to determine whether a similar relationship exists with respect to interleukin 1-beta (IL-1 beta), whose receptor activation in the medial hypothalamus potentiates defensive rage. Thus, the present study identified the effects of administration of IL-1 beta into the PAG upon defensive rage elicited from the medial hypothalamus. Microinjections of IL-1 beta into the dorsal PAG significantly facilitated defensive rage behavior elicited from the medial hypothalamus in a dose and time dependent manner. In addition, the facilitative effects of IL-1 beta were blocked by pre-treatment with anti-IL-1 beta receptor antibody, while IL-1 beta administration into the PAG had no effect upon predatory attack elicited from the lateral hypothalamus. The findings further demonstrated that IL-1 beta's effects were mediated through 5-HT(2) receptors since pretreatment with a 5-HT(2C) receptors antagonist blocked the facilitating effects of IL-1 beta. An extensive pattern of labeling of IL-1 beta and 5-HT(2C) receptors in the dorsal PAG supported these findings. The present study demonstrates that IL-beta in the dorsal PAG, similar to the medial hypothalamus, potentiates defensive rage behavior and is mediated through a 5-HT(2C) receptor mechanism.


Subject(s)
Interleukin-1beta/physiology , Neuroimmunomodulation/physiology , Periaqueductal Gray/physiology , Rage/physiology , Receptor, Serotonin, 5-HT2C/physiology , Animals , Antibodies/pharmacology , Behavior, Animal/physiology , Cats , Ergolines/pharmacology , Female , Hypothalamic Area, Lateral/physiology , Hypothalamus, Middle/physiology , Interleukin-1beta/pharmacology , Microinjections , Periaqueductal Gray/drug effects , Predatory Behavior/physiology , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/pharmacology , Tachykinins/pharmacology
14.
Curr Neuropharmacol ; 5(2): 135-47, 2007.
Article in English | MEDLINE | ID: mdl-18615178

ABSTRACT

Violence and aggression are major causes of death and injury, thus constituting primary public health problems throughout much of the world costing billions of dollars to society. The present review relates our understanding of the neurobiology of aggression and rage to pharmacological treatment strategies that have been utilized and those which may be applied in the future. Knowledge of the neural mechanisms governing aggression and rage is derived from studies in cat and rodents. The primary brain structures involved in the expression of rage behavior include the hypothalamus and midbrain periaqueductal gray. Limbic structures, which include amygdala, hippocampal formation, septal area, prefrontal cortex and anterior cingulate gyrus serve important modulating functions. Excitatory neurotransmitters that potentiate rage behavior include excitatory amino acids, substance P, catecholamines, cholecystokinin, vasopressin, and serotonin that act through 5-HT(2) receptors. Inhibitory neurotransmitters include GABA, enkephalins, and serotonin that act through 5-HT(1) receptors. Recent studies have demonstrated that brain cytokines, including IL-1beta and IL-2, powerfully modulate rage behavior. IL-1-beta exerts its actions by acting through 5-HT(2) receptors, while IL-2 acts through GABAA or NK(1) receptors. Pharmacological treatment strategies utilized for control of violent behavior have met with varying degrees of success. The most common approach has been to apply serotonergic compounds. Others included the application of antipsychotic, GABAergic (anti-epileptic) and dopaminergic drugs. Present and futures studies on the neurobiology of aggression may provide the basis for new and novel treatment strategies for the control of aggression and violence as well as the continuation of existing pharmacological approaches.

15.
Brain Behav Immun ; 20(6): 507-14, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16938427

ABSTRACT

Recent studies have suggested an important relationship linking cytokines, immunity and aggressive behavior. Clinical reports describe increasing levels of hostility, anger, and irritability in patients who receive cytokine immunotherapy, and there are reports of a positive correlation between cytokine levels and aggressive behavior in non-patient populations. On the basis of these reports and others describing the presence or actions of different cytokines in regions of the brain associated with aggressive behavior, our laboratory embarked upon a program of research designed to identify and characterize the role of IL-1 and IL-2 in the hypothalamus and midbrain periaqueductal gray (PAG)--two regions functionally linked through reciprocal anatomical connections--in the regulation of feline defensive rage. A paradigm involved cytokine microinjections into either medial hypothalamus and elicitation of defensive rage behavior from the PAG or vice versa. These studies have revealed that both cytokines have potent effects in modulating defensive rage behavior. With respect to IL-1, this cytokine facilitates defensive rage when microinjected into either the medial hypothalamus or PAG and these potentiating effects are mediated through 5-HT2 receptors. In contrast, the effects of IL-2 are dependent upon the anatomical locus. IL-2 microinjected into the medial hypothalamus suppresses defensive rage and this suppression is mediated through GABA(A) receptors, while microinjections of IL-2 in the PAG potentiate defensive rage, in which these effects are mediated through NK-1 receptors. Present research is designed to further delineate the roles of cytokines in aggressive behavior and to begin to unravel the possible signaling pathways involved this process.


Subject(s)
Aggression/physiology , Hypothalamus/immunology , Interleukin-1/physiology , Interleukin-2/physiology , Periaqueductal Gray/immunology , Rage/physiology , Animals , Cats , Neural Pathways/immunology , Neurobiology
16.
Neurobiol Dis ; 21(1): 127-37, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16084731

ABSTRACT

The present study sought to determine the effects of long-term kindled seizures of the basal amygdala upon immune function in rat, utilizing the thymus, as a principal target for study. Histopathology from kindled Sprague-Dawley rats revealed the presence of epithelial cell thymoma in 70% of these rats. The results revealed an increased rate of apoptosis and proliferation in thymic epithelial cells. Analysis of thymocytes indicated a decrease in the ratio of CD4 to CD8 positive T cells and reduced proliferative response to T-cell mitogens. To determine whether these effects were mediated through the sympathetic nervous system, animals were treated with guanethidine, which blocked the development of epithelial cell thymomas, while mifepristone treatment, employed to determine the possible role of the hypothalamic-pituitary axis, was ineffective in attenuating thymoma development. Thus, the present study demonstrated that functional and pathological changes in the thymus during kindled seizures are mediated through the sympathetic nervous system.


Subject(s)
Amygdala/physiopathology , Kindling, Neurologic/physiology , Sympathetic Nervous System/physiopathology , Thymus Gland/pathology , Thymus Gland/physiopathology , Animals , Antibodies , Apoptosis/drug effects , Cell Division , Epilepsy/physiopathology , Epithelial Cells/pathology , Female , Guanethidine/pharmacology , Hormone Antagonists/pharmacology , In Situ Nick-End Labeling , Keratins/immunology , Keratins/metabolism , Mifepristone/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic/metabolism , Sympatholytics/pharmacology , T-Lymphocytes/pathology , Thymoma/drug therapy , Thymoma/etiology , Thymoma/pathology , Thymus Gland/innervation , Thymus Neoplasms/drug therapy , Thymus Neoplasms/etiology , Thymus Neoplasms/pathology
17.
Behav Brain Res ; 167(2): 251-60, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16242788

ABSTRACT

Feline defensive rage is a form of aggression occurring in nature in response to a threatening condition and is elicited under laboratory conditions by electrical stimulation of the medial hypothalamus or midbrain periaqueductal gray (PAG). Since it has recently been shown that cytokines can modulate neurotransmitter release, the present study was designed to determine the effects of administration of interleukin 2 (IL-2) into the PAG upon defensive rage elicited from the medial hypothalamus. Microinjections of relatively low doses of IL-2 into the dorsal PAG significantly facilitated defensive rage behavior elicited from the medial hypothalamus. The specificity of this phenomenon was supported by the following findings: (1) IL-2 induced effects were dose- and time-dependent, (2) the facilitative effects of IL-2 could be completely blocked by pre-treatment of the injection site with either anti-IL-2 or anti-IL-2 receptor antibody and (3) IL-2 administration into the PAG showed no effect upon another form of aggression, namely predatory attack, elicited from the lateral hypothalamus. The findings further demonstrated that the effects of IL-2 were mediated by an NK(1) receptor mechanism since pre-treatment of the PAG with an NK(1) receptor antagonist completely blocked the facilitating effects of IL-2. Immunocytochemical observations supported these findings by demonstrating an extensive pattern of labeling of IL-2Ralpha in the dorsal PAG. The present study thus demonstrates that IL-2 in the dorsal PAG potentiates defensive rage behavior and is mediated through an NK(1) receptor mechanism.


Subject(s)
Periaqueductal Gray/physiology , Rage/physiology , Receptors, Interleukin-2/physiology , Receptors, Neurokinin-1/physiology , Aggression/physiology , Analysis of Variance , Animals , Cats , Cytokines/administration & dosage , Cytokines/physiology , Dose-Response Relationship, Drug , Electric Stimulation , Female , Hypothalamus/physiology , Interleukin-2/administration & dosage , Interleukin-2/physiology , Microinjections , Periaqueductal Gray/drug effects , Rage/drug effects , Receptors, Neurokinin-1/drug effects , Statistics, Nonparametric
18.
Epilepsy Res ; 65(3): 169-78, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16040233

ABSTRACT

Recent studies conducted in our laboratory have demonstrated marked increases in both serum leptin levels and colony numbers in bone marrow progenitor cells following long-term kindled seizures in rats. The present study sought to determine whether such changes in hematopoietic functions following kindling are linked to increased serum leptin levels. Kindled stage V seizures were induced for 30 days in Sprague-Dawley rats by stimulation of the basal complex of amygdala. The results revealed colony numbers in colony forming units-granulocyte/macrophage (CFU-GM) cultures from kindled rats increased significantly, an effect that was blocked by the presence of an anti-leptin antibody. The results further demonstrated that the addition of serum obtained from kindled rats to CFU-GM cultures from control rats significantly increased the numbers of colonies relative to non-serum added cultures. Moreover, the proliferative effects of serum from kindled rats were also blocked by adding an anti-leptin antibody. These findings were confirmed from the observations that the long isoform of the leptin receptor, which is capable of signal transduction, was present only in kindled, but not in control rats. Thus, the results provide evidence that the hematopoietic changes observed following long-term kindling are directly associated with elevated serum leptin levels.


Subject(s)
Hematopoiesis/physiology , Kindling, Neurologic , Leptin/blood , Seizures/blood , Analysis of Variance , Animals , Antibodies/pharmacology , Cell Count , Colony-Forming Units Assay/methods , Female , Hematopoiesis/drug effects , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Kindling, Neurologic/drug effects , Leptin/immunology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Leptin , Reverse Transcriptase Polymerase Chain Reaction/methods , Seizures/physiopathology , Time Factors
19.
Brain Res ; 994(1): 55-66, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14642448

ABSTRACT

This study utilized anatomical and behavioral-pharmacological methods to determine the role of NK(1)-Substance P receptors in the midbrain periaqueductal gray (PAG) in defensive rage behavior in cats. For behavioral pharmacological experiments, monopolar stimulating electrodes were implanted in the medial hypothalamus for elicitation of defensive rage behavior and cannula-electrodes were implanted in the PAG for microinjections of receptor compounds. Microinjections of the NMDA antagonist, AP-7 (2 nmol), into the dorsal PAG blocked defensive rage elicited by medial hypothalamic stimulation, thus establishing the PAG as a synaptic region that receives hypothalamic inputs linked to defensive rage behavior. Microinjections of the NK(1) agonist, GR73632, into the same injection sites facilitated defensive rage in a dose-dependent manner, and also induced spontaneous hissing in five cats. The effects of GR73632 were reduced by pretreatment of the PAG with the NK(1) antagonist, GR82334 (16 nmol), microinjected into the same sites. Microinjections of GR73632 (8 nmol) into the PAG also suppressed predatory attack elicited by stimulation of the lateral hypothalamus. Immunohistochemical methods utilized to detect Substance P and Fos immunoreactivity revealed that neurons in the PAG activated after defensive rage-inducing medial hypothalamic stimulation lie in the same region as Substance-P-immunoreactive processes. Fos immunoreactivity was highest in the dorsomedial aspect of the rostral PAG after medial hypothalamic stimulation. Cats that were unstimulated or that exhibited predatory attack after lateral hypothalamic stimulation had low c-fos expression levels in the PAG. Substance P immunoreactivity was high throughout the dorsal PAG. The results indicate that NK(1) receptors in the PAG potentiate defensive rage and suppress predatory aggression in the cat.


Subject(s)
Mesencephalon/metabolism , Periaqueductal Gray/metabolism , Predatory Behavior/physiology , Rage/physiology , Receptors, Neurokinin-1/metabolism , Animals , Cats , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Mesencephalon/drug effects , Neurokinin-1 Receptor Antagonists , Periaqueductal Gray/drug effects , Predatory Behavior/drug effects , Rage/drug effects , Receptors, Neurokinin-1/agonists
20.
Epilepsy Res ; 54(2-3): 209-19, 2003 May.
Article in English | MEDLINE | ID: mdl-12837572

ABSTRACT

OBJECTIVE: Studies conducted in epilepsy patients and experimental animals have suggested a linkage between seizure activity and alterations in immune functions. However, little is known about the underlying mechanisms. The present study sought to determine whether chronic seizures result in changes in hematopoietic functions that contribute to alterations in immune function. MATERIALS AND METHODS: Sprague-Dawley rats were implanted with electrodes in the basal amygdala or frontal cortex for induction of focal seizures by kindling. After inducing stage 5 seizures for 30 days, rats were sacrificed and assays for colony-forming units granulocyte/macrophage (CFU-GM) were performed to study progenitor cell functions. Long-term culture-initiating culture (LTC-IC) assays were employed to determine the effects of kindling upon bone marrow stroma. A Western blot for caspase-3 and CFU-GM assays from peripheral blood were used to determine the cause of reduced cellularity of bone marrow. RESULTS: Kindled seizures of the basal amygdala resulted in decreases in bone marrow cellularity and hyperproliferation of colony-forming cells in peripheral blood and bone marrow. Modified LTC-IC assays, where co-cultures of bone marrow cells and stroma from experimental animals were employed, revealed that hyperproliferation of progenitor cells was not associated with alterations in stromal functions. The changes observed in this study were associated with seizure foci in the basal amygdaloid complex but not the frontal cortex. CONCLUSION: Kindled seizures of the basal amygdala induce hyperproliferation of bone marrow progenitor cells, suggesting that alterations in immunological functions observed following seizure activity may be due to changes in hematopoietic functions. Such changes appear to be site specific within the brain.


Subject(s)
Hematopoiesis , Kindling, Neurologic/physiology , Seizures/blood , Seizures/physiopathology , Animals , Blotting, Western , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , Caspase 3 , Caspases/analysis , Cell Division/physiology , Clone Cells , Colony-Forming Units Assay , Female , Femur , Hematopoiesis/physiology , Leukocyte Count , Rats , Rats, Sprague-Dawley , Seizures/enzymology , Stem Cells/pathology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...