Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 112(29): 9118-22, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26150480

ABSTRACT

Understanding the heritability of neural systems linked to psychopathology is not sufficient to implicate them as intergenerational neural mediators. By closely examining how individual differences in neural phenotypes and psychopathology cosegregate as they fall through the family tree, we can identify the brain systems that underlie the parent-to-child transmission of psychopathology. Although research has identified genes and neural circuits that contribute to the risk of developing anxiety and depression, the specific neural systems that mediate the inborn risk for these debilitating disorders remain unknown. In a sample of 592 young rhesus monkeys that are part of an extended multigenerational pedigree, we demonstrate that metabolism within a tripartite prefrontal-limbic-midbrain circuit mediates some of the inborn risk for developing anxiety and depression. Importantly, although brain volume is highly heritable early in life, it is brain metabolism-not brain structure-that is the critical intermediary between genetics and the childhood risk to develop stress-related psychopathology.


Subject(s)
Anxiety/physiopathology , Family Characteristics , Inheritance Patterns , Neurons/physiology , Temperament , Animals , Brain/metabolism , Brain/physiopathology , Macaca mulatta
2.
Biol Psychiatry ; 76(11): 850-7, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24342924

ABSTRACT

BACKGROUND: Anxious temperament (AT) is identifiable early in life and predicts the later development of anxiety disorders and depression. Neuropeptide Y (NPY) is a putative endogenous anxiolytic neurotransmitter that adaptively regulates responses to stress and might confer resilience to stress-related psychopathology. With a well-validated nonhuman primate model of AT, we examined expression of the NPY system in the central nucleus (Ce) of the amygdala, a critical neural substrate for extreme anxiety. METHODS: In 24 young rhesus monkeys, we measured Ce messenger RNA (mRNA) levels of all members of the NPY system that are detectable in the Ce with quantitative real time polymerase chain reaction. We then examined the relationship between these mRNA levels and both AT expression and brain metabolism. RESULTS: Lower mRNA levels of neuropeptide Y receptor 1 (NPY1R) and NPY5R but not NPY or NPY2R in the Ce predicted elevated AT; mRNA levels for NPY1R and NPY5R in the motor cortex were not related to AT. In situ hybridization analysis provided for the first time a detailed description of NPY1R and NPY5R mRNA distribution in the rhesus amygdala and associated regions. Lastly, mRNA levels for these two receptors in the Ce predicted metabolic activity in several regions that have the capacity to regulate the Ce. CONCLUSIONS: Decreased NPY signaling in the Ce might contribute to the altered metabolic activity that is a component of the neural substrate underlying AT. This suggests that enhancement of NPY signaling might reduce the risk to develop psychopathology.


Subject(s)
Anxiety/metabolism , Brain/metabolism , Central Amygdaloid Nucleus/metabolism , Receptors, Neuropeptide Y/genetics , Animals , Anxiety/physiopathology , Brain/physiopathology , Central Amygdaloid Nucleus/physiopathology , Gene Expression , Macaca mulatta , RNA, Messenger/metabolism , Receptors, Neuropeptide Y/physiology
3.
Proc Natl Acad Sci U S A ; 110(15): 6145-50, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23538303

ABSTRACT

Children with an anxious temperament (AT) are at risk for developing psychiatric disorders along the internalizing spectrum, including anxiety and depression. Like these disorders, AT is a multidimensional phenotype and children with extreme anxiety show varying mixtures of physiological, behavioral, and other symptoms. Using a well-validated juvenile monkey model of AT, we addressed the degree to which this phenotypic heterogeneity reflects fundamental differences or similarities in the underlying neurobiology. The rhesus macaque is optimal for studying AT because children and young monkeys express the anxious phenotype in similar ways and have similar neurobiology. Fluorodeoxyglucose (FDG)-positron emission tomography (FDG-PET) in 238 freely behaving monkeys identified brain regions where metabolism predicted variation in three dimensions of the AT phenotype: hypothalamic-pituitary-adrenal (HPA) activity, freezing behavior, and expressive vocalizations. We distinguished brain regions that predicted all three dimensions of the phenotype from those that selectively predicted a single dimension. Elevated activity in the central nucleus of the amygdala and the anterior hippocampus was consistently found across individuals with different presentations of AT. In contrast, elevated activity in the lateral anterior hippocampus was selective to individuals with high levels of HPA activity, and decreased activity in the motor cortex (M1) was selective to those with high levels of freezing behavior. Furthermore, activity in these phenotype-selective regions mediated relations between amygdala metabolism and different expressions of anxiety. These findings provide a framework for understanding the mechanisms that lead to heterogeneity in the clinical presentation of internalizing disorders and set the stage for developing improved interventions.


Subject(s)
Anxiety/pathology , Brain Mapping/methods , Brain/pathology , Amygdala/physiology , Animals , Anxiety/metabolism , Brain/metabolism , Depression/metabolism , False Positive Reactions , Female , Hippocampus/physiology , Hydrocortisone/blood , Hydrocortisone/metabolism , Macaca mulatta , Male , Models, Animal , Models, Neurological , Neuroimaging/methods , Phenotype , Radioimmunoassay , Time Factors
4.
Proc Natl Acad Sci U S A ; 109(44): 18108-13, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23071305

ABSTRACT

Children with anxious temperament (AT) are particularly sensitive to new social experiences and have increased risk for developing anxiety and depression. The young rhesus monkey is optimal for studying the origin of human AT because it shares with humans the genetic, neural, and phenotypic underpinnings of complex social and emotional functioning. In vivo imaging in young monkeys demonstrated that central nucleus of the amygdala (Ce) metabolism is relatively stable across development and predicts AT. Transcriptome-wide gene expression, which reflects combined genetic and environmental influences, was assessed within the Ce. Results support a maladaptive neurodevelopmental hypothesis linking decreased amygdala neuroplasticity to early-life dispositional anxiety. For example, high AT individuals had decreased mRNA expression of neurotrophic tyrosine kinase, receptor, type 3 (NTRK3). Moreover, variation in Ce NTRK3 expression was inversely correlated with Ce metabolism and other AT-substrates. These data suggest that altered amygdala neuroplasticity may play a role the early dispositional risk to develop anxiety and depression.


Subject(s)
Amygdala/metabolism , Anxiety/genetics , Gene Expression , Animals , Macaca mulatta , Male
5.
Neuroimage ; 61(4): 1059-66, 2012 Jul 16.
Article in English | MEDLINE | ID: mdl-22465841

ABSTRACT

Neuroanatomists posit that the central nucleus of the amygdala (Ce) and bed nucleus of the stria terminalis (BST) comprise two major nodes of a macrostructural forebrain entity termed the extended amygdala. The extended amygdala is thought to play a critical role in adaptive motivational behavior and is implicated in the pathophysiology of maladaptive fear and anxiety. Resting functional connectivity of the Ce was examined in 107 young anesthetized rhesus monkeys and 105 young humans using standard resting-state functional magnetic resonance imaging (fMRI) methods to assess temporal correlations across the brain. The data expand the neuroanatomical concept of the extended amygdala by finding, in both species, highly significant functional coupling between the Ce and the BST. These results support the use of in vivo functional imaging methods in nonhuman and human primates to probe the functional anatomy of major brain networks such as the extended amygdala.


Subject(s)
Amygdala/anatomy & histology , Amygdala/physiology , Neural Pathways/anatomy & histology , Neural Pathways/physiology , Adolescent , Animals , Anxiety/physiopathology , Child , Fear/physiology , Female , Humans , Image Processing, Computer-Assisted , Macaca mulatta , Magnetic Resonance Imaging , Male
6.
Neuroimage ; 59(1): 306-18, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-21803162

ABSTRACT

Diffusion tensor imaging (DTI) is a powerful and noninvasive imaging method for characterizing tissue microstructure and white matter organization in the brain. While it has been applied extensively in research studies of the human brain, DTI studies of non-human primates have been performed only recently. The growing application of DTI in rhesus monkey studies would significantly benefit from a standardized framework to compare findings across different studies. A very common strategy for image analysis is to spatially normalize (co-register) the individual scans to a representative template space. This paper presents the development of a DTI brain template, UWRMAC-DTI271, for adolescent Rhesus Macaque (Macaca mulatta) monkeys. The template was generated from 271 rhesus monkeys, collected as part of a unique brain imaging genetics study. It is the largest number of animals ever used to generate a computational brain template, which enables the generation of a template that has high image quality and accounts for variability in the species. The quality of the template is further ensured with the use of DTI-TK, a well-tested and high-performance DTI spatial normalization method in human studies. We demonstrated its efficacy in monkey studies for the first time by comparing it to other commonly used scalar-methods for DTI normalization. It is anticipated that this template will play an important role in facilitating cross-site voxelwise DTI analyses in Rhesus Macaques. Such analyses are crucial in investigating the role of white matter structure in brain function, development, and other psychopathological disorders for which there are well-validated non-human primate models.


Subject(s)
Brain Mapping , Brain/anatomy & histology , Diffusion Magnetic Resonance Imaging , Macaca mulatta/anatomy & histology , Animals , Image Processing, Computer-Assisted
7.
BMC Genomics ; 12: 311, 2011 Jun 13.
Article in English | MEDLINE | ID: mdl-21668978

ABSTRACT

BACKGROUND: Rhesus macaques are the most widely utilized nonhuman primate model in biomedical research. Previous efforts have validated fewer than 900 single nucleotide polymorphisms (SNPs) in this species, which limits opportunities for genetic studies related to health and disease. Extensive information about SNPs and other genetic variation in rhesus macaques would facilitate valuable genetic analyses, as well as provide markers for genome-wide linkage analysis and the genetic management of captive breeding colonies. RESULTS: We used the available rhesus macaque draft genome sequence, new sequence data from unrelated individuals and existing published sequence data to create a genome-wide SNP resource for Indian-origin rhesus monkeys. The original reference animal and two additional Indian-origin individuals were resequenced to low coverage using SOLiD™ sequencing. We then used three strategies to validate SNPs: comparison of potential SNPs found in the same individual using two different sequencing chemistries, and comparison of potential SNPs in different individuals identified with either the same or different sequencing chemistries. Our approach validated approximately 3 million SNPs distributed across the genome. Preliminary analysis of SNP annotations suggests that a substantial number of these macaque SNPs may have functional effects. More than 700 non-synonymous SNPs were scored by Polyphen-2 as either possibly or probably damaging to protein function and these variants now constitute potential models for studying functional genetic variation relevant to human physiology and disease. CONCLUSIONS: Resequencing of a small number of animals identified greater than 3 million SNPs. This provides a significant new information resource for rhesus macaques, an important research animal. The data also suggests that overall genetic variation is high in this species. We identified many potentially damaging non-synonymous coding SNPs, providing new opportunities to identify rhesus models for human disease.


Subject(s)
Genetic Variation , Macaca mulatta/genetics , Polymorphism, Single Nucleotide , Animals , India , Sequence Analysis, DNA , Species Specificity
8.
Nature ; 466(7308): 864-8, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20703306

ABSTRACT

Anxious temperament (AT) in human and non-human primates is a trait-like phenotype evident early in life that is characterized by increased behavioural and physiological reactivity to mildly threatening stimuli. Studies in children demonstrate that AT is an important risk factor for the later development of anxiety disorders, depression and comorbid substance abuse. Despite its importance as an early predictor of psychopathology, little is known about the factors that predispose vulnerable children to develop AT and the brain systems that underlie its expression. To characterize the neural circuitry associated with AT and the extent to which the function of this circuit is heritable, we studied a large sample of rhesus monkeys phenotyped for AT. Using 238 young monkeys from a multigenerational single-family pedigree, we simultaneously assessed brain metabolic activity and AT while monkeys were exposed to the relevant ethological condition that elicits the phenotype. High-resolution (18)F-labelled deoxyglucose positron-emission tomography (FDG-PET) was selected as the imaging modality because it provides semi-quantitative indices of absolute glucose metabolic rate, allows for simultaneous measurement of behaviour and brain activity, and has a time course suited for assessing temperament-associated sustained brain responses. Here we demonstrate that the central nucleus region of the amygdala and the anterior hippocampus are key components of the neural circuit predictive of AT. We also show significant heritability of the AT phenotype by using quantitative genetic analysis. Additionally, using voxelwise analyses, we reveal significant heritability of metabolic activity in AT-associated hippocampal regions. However, activity in the amygdala region predictive of AT is not significantly heritable. Furthermore, the heritabilities of the hippocampal and amygdala regions significantly differ from each other. Even though these structures are closely linked, the results suggest differential influences of genes and environment on how these brain regions mediate AT and the ongoing risk of developing anxiety and depression.


Subject(s)
Amygdala/metabolism , Anxiety/genetics , Anxiety/physiopathology , Genetic Predisposition to Disease/genetics , Heredity , Hippocampus/metabolism , Temperament/physiology , Animals , Depression/genetics , Female , Freezing Reaction, Cataleptic , Glucose/metabolism , Macaca mulatta/genetics , Macaca mulatta/physiology , Male , Models, Animal , Neural Pathways/physiology , Pedigree , Phenotype , Positron-Emission Tomography , Stress, Psychological , Temporal Lobe/metabolism , Vocalization, Animal
9.
J Neurosci ; 30(20): 7023-7, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484644

ABSTRACT

In children, behavioral inhibition (BI) in response to potential threat predicts the development of anxiety and affective disorders, and primate lesion studies suggest involvement of the orbitofrontal cortex (OFC) in mediating BI. Lesion studies are essential for establishing causality in brain-behavior relationships, but should be interpreted cautiously because the impact of a discrete lesion on a complex neural circuit extends beyond the lesion location. Complementary functional imaging methods assessing how lesions influence other parts of the circuit can aid in precisely understanding how lesions affect behavior. Using this combination of approaches in monkeys, we found that OFC lesions concomitantly alter BI and metabolism in the bed nucleus of stria terminalis (BNST) region and that individual differences in BNST activity predict BI. Thus it appears that an important function of the OFC in response to threat is to modulate the BNST, which may more directly influence the expression of BI.


Subject(s)
Anxiety/pathology , Brain Injuries/complications , Brain Injuries/pathology , Prefrontal Cortex/pathology , Septal Nuclei/physiopathology , Amygdala/diagnostic imaging , Amygdala/physiopathology , Animals , Anxiety/diagnostic imaging , Behavior, Animal/physiology , Brain Mapping , Fluorodeoxyglucose F18 , Freezing Reaction, Cataleptic/physiology , Macaca mulatta , Magnetic Resonance Imaging/methods , Male , Positron-Emission Tomography/methods , Septal Nuclei/diagnostic imaging
10.
Biol Psychiatry ; 67(2): 175-81, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19846063

ABSTRACT

BACKGROUND: Hypothalamic-pituitary-adrenal (HPA) system activation is adaptive in response to stress, and HPA dysregulation occurs in stress-related psychopathology. It is important to understand the mechanisms that modulate HPA output, yet few studies have addressed the neural circuitry associated with HPA regulation in primates and humans. Using high-resolution F-18-fluorodeoxyglucose positron emission tomography (FDG-PET) in rhesus monkeys, we assessed the relation between individual differences in brain activity and HPA function across multiple contexts that varied in stressfulness. METHODS: Using a logical AND conjunctions analysis, we assessed cortisol and brain metabolic activity with FDG-PET in 35 adolescent rhesus monkeys exposed to two threat and two home-cage conditions. To test the robustness of our findings, we used similar methods in an archival data set. In this data set, brain metabolic activity and cortisol were assessed in 17 adolescent male rhesus monkeys that were exposed to three stress-related contexts. RESULTS: Results from the two studies revealed that subgenual prefrontal cortex (PFC) metabolism (Brodmann's area 25/24) consistently predicted individual differences in plasma cortisol concentrations regardless of the context in which brain activity and cortisol were assessed. CONCLUSIONS: These findings suggest that activation in subgenual PFC may be related to HPA output across a variety of contexts (including familiar settings and novel or threatening situations). Individuals prone to elevated subgenual PFC activity across multiple contexts may be individuals who consistently show heightened cortisol and may be at risk for stress-related HPA dysregulation.


Subject(s)
Brain Mapping , Hypothalamo-Hypophyseal System/physiopathology , Individuality , Pituitary-Adrenal System/physiopathology , Prefrontal Cortex/physiopathology , Stress, Psychological/pathology , Animals , Brain/metabolism , Disease Models, Animal , Female , Fluorodeoxyglucose F18 , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/diagnostic imaging , Immunoassay/methods , Logistic Models , Macaca mulatta , Magnetic Resonance Imaging , Male , Pituitary-Adrenal System/diagnostic imaging , Positron-Emission Tomography/methods , Predictive Value of Tests , Prefrontal Cortex/diagnostic imaging , Probability , Social Isolation/psychology , Stress, Psychological/diagnostic imaging
11.
J Neurosci ; 29(32): 9961-6, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19675230

ABSTRACT

The serotonin transporter (5-HTT) plays a critical role in regulating serotonergic neurotransmission and is implicated in the pathophysiology of anxiety and affective disorders. Positron emission tomography scans using [(11)C]DASB [(11)C]-3-amino-4-(2-dimethylaminomethylphenylsulfanyl)-benzonitrile] to measure 5-HTT availability (an index of receptor density and binding) were performed in 34 rhesus monkeys in which the relationship between regional brain glucose metabolism and anxious temperament was previously established. 5-HTT availability in the amygdalohippocampal area and bed nucleus of the stria terminalis correlated positively with individual differences in a behavioral and neuroendocrine composite of anxious temperament. 5-HTT availability also correlated positively with stress-induced metabolic activity within these regions. Collectively, these findings suggest that serotonergic modulation of neuronal excitability in the neural circuitry associated with anxiety mediates the developmental risk for affect-related psychopathology.


Subject(s)
Amygdala/physiology , Anxiety/physiopathology , Glucose/metabolism , Septal Nuclei/physiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Temperament/physiology , Amygdala/diagnostic imaging , Aniline Compounds , Animals , Anxiety/diagnostic imaging , Behavior, Animal , Female , Fluorodeoxyglucose F18 , Linear Models , Macaca mulatta , Male , Positron-Emission Tomography , Septal Nuclei/diagnostic imaging , Sulfides
12.
PLoS One ; 3(7): e2570, 2008 Jul 02.
Article in English | MEDLINE | ID: mdl-18596957

ABSTRACT

Early theorists (Freud and Darwin) speculated that extremely shy children, or those with anxious temperament, were likely to have anxiety problems as adults. More recent studies demonstrate that these children have heightened responses to potentially threatening situations reacting with intense defensive responses that are characterized by behavioral inhibition (BI) (inhibited motor behavior and decreased vocalizations) and physiological arousal. Confirming the earlier impressions, data now demonstrate that children with this disposition are at increased risk to develop anxiety, depression, and comorbid substance abuse. Additional key features of anxious temperament are that it appears at a young age, it is a stable characteristic of individuals, and even in non-threatening environments it is associated with increased psychic anxiety and somatic tension. To understand the neural underpinnings of anxious temperament, we performed imaging studies with 18-fluoro-deoxyglucose (FDG) high-resolution Positron Emission Tomography (PET) in young rhesus monkeys. Rhesus monkeys were used because they provide a well validated model of anxious temperament for studies that cannot be performed in human children. Imaging the same animal in stressful and secure contexts, we examined the relation between regional metabolic brain activity and a trait-like measure of anxious temperament that encompasses measures of BI and pituitary-adrenal reactivity. Regardless of context, results demonstrated a trait-like pattern of brain activity (amygdala, bed nucleus of stria terminalis, hippocampus, and periaqueductal gray) that is predictive of individual phenotypic differences. Importantly, individuals with extreme anxious temperament also displayed increased activity of this circuit when assessed in the security of their home environment. These findings suggest that increased activity of this circuit early in life mediates the childhood temperamental risk to develop anxiety and depression. In addition, the findings provide an explanation for why individuals with anxious temperament have difficulty relaxing in environments that others perceive as non-stressful.


Subject(s)
Anxiety , Behavior, Animal/physiology , Brain/physiology , Temperament/physiology , Animals , Female , Humans , Macaca mulatta , Male , Positron-Emission Tomography
13.
Hum Gene Ther ; 19(7): 690-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18576918

ABSTRACT

Gliomas have a dismal prognosis, with the median survival of patients with the most common histology, glioblastoma multiforme, being only 12-15 months. Development of novel therapeutic agents is urgently needed. We have previously demonstrated that oncolytic measles virus strains derived from the Edmonston vaccine lineage have significant antitumor activity against gliomas [Phuong, L.K., Allen, C., Peng, K.W., Giannini, C., Greiner, S., Teneyck, C.J., Mishra, P.K., Macura, S.I., Russell, S.J., Galanis, E.C. (2003). Cancer. Res. 63, 2462-2469]. MV-CEA is an Edmonston vaccine lineage measles virus strain engineered to express the marker peptide carcinoembryonic antigen (CEA): CEA levels can serve as a correlate of viral gene expression. In support of a phase I clinical trial of intratumoral and resection cavity administration of MV-CEA to patients with recurrent gliomas, we assessed the neurotoxicity of MV-CEA in adult immune male rhesus macaques (Macaca mulatta). The animals ' immune status and administration schedule mimicked the trial population and proposed administration schema. Macaca mulatta represents the prototype animal species for assessment of measles neurotoxicity. The animals were stereotactically administered either vehicle (n = 1) or MV-CEA at 2 x 10(5)or 2 x 10(6) TCID(50) (each, n = 2) in the right frontal lobe in two injections on days 1 and 5. Macaques were closely monitored clinically for neurotoxicity. Body weight, temperature, complete blood count, CEA, clinical chemistries, coagulation, complement levels, immunoglobulin, measles antibody titers, viremia, and shedding (buccal swabs) were tested at multiple time points. Furthermore, cisterna magna spinal taps were performed on day 9 and 1 year after the first viral dose administration, and samples were analyzed for protein, glucose, cell differential, and presence of MV-CEA. Magnetic resonance imaging (MRI) was performed between 4 and 5 months after article administration to assess for subclinical neurotoxicity. To date, 36+ months from study initiation there has been no clinical or biochemical evidence of toxicity, including lack of neurological symptoms, fever, or other systemic symptoms and lack of immunosuppression. Quantitative RT-PCR analysis of blood, buccal swabs, and cerebrospinal fluid (CSF) was negative for MV-CEA at all time points, with the exception of viral genome deletion in the blood of one asymptomatic animal at the 2 x 10(6) TCID(50) dose level on day 85. Vero cell overlays of CSF cells and supernatant were negative for viral recovery. There was no detection of CEA in serum or CSF at any time point. MRI scans were negative for imaging abnormalities and showed no evidence of encephalitis. Our results support the safety of CNS administration of MV-CEA in glioma patients. A clinical trial of intratumoral and resection cavity administration of MV-CEA in patients with recurrent glioblastoma multiforme is currently ongoing.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines , Carcinoembryonic Antigen/metabolism , Genetic Vectors , Glioma/therapy , Measles virus , Animals , Brain/virology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Carcinoembryonic Antigen/genetics , Chlorocebus aethiops , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Cytopathogenic Effect, Viral , Drug Administration Routes , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Glioma/genetics , Glioma/pathology , Glioma/prevention & control , Humans , Macaca mulatta , Male , Measles virus/genetics , Measles virus/pathogenicity , Recurrence , Treatment Outcome , Vero Cells
14.
PLoS One ; 3(3): e1751, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18347739

ABSTRACT

Functional MRI resting state and connectivity studies of brain focus on neural fluctuations at low frequencies which share power with physiological fluctuations originating from lung and heart. Due to the lack of automated software to process physiological signals collected at high magnetic fields, a gap exists in the processing pathway between the acquisition of physiological data and its use in fMRI software for both physiological noise correction and functional analyses of brain activation and connectivity. To fill this gap, we developed an open source, physiological signal processing program, called PhysioNoise, in the python language. We tested its automated processing algorithms and dynamic signal visualization on resting monkey cardiac and respiratory waveforms. PhysioNoise consistently identifies physiological fluctuations for fMRI noise correction and also generates covariates for subsequent analyses of brain activation and connectivity.


Subject(s)
Magnetic Resonance Imaging/methods , Algorithms , Heart/physiology , Respiration
15.
Biol Psychiatry ; 62(10): 1134-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17643397

ABSTRACT

BACKGROUND: Excessive behavioral inhibition during childhood marks anxious temperament and is a risk factor for the development of anxiety and affective disorders. Studies in nonhuman primates can provide important information related to the expression of this risk factor, since threat-induced freezing in rhesus monkeys is a trait-like characteristic analogous to human behavioral inhibition. The orbitofrontal cortex (OFC) and amygdala are part of a circuit involved in the processing of emotions and associated physiological responses. Earlier work demonstrated involvement of the primate central nucleus of the amygdala (CeA) in mediating anxious temperament. This study assessed the role of the primate OFC in mediating anxious temperament and its involvement in fear responses. METHODS: Twelve adolescent rhesus monkeys were studied (six lesion and six control monkeys). Lesions were targeted at regions of the OFC that are most interconnected with the amygdala. Behavior and physiological parameters were assessed before and after the lesions. RESULTS: The OFC lesions significantly decreased threat-induced freezing and marginally decreased fearful responses to a snake. The lesions also resulted in a leftward shift in frontal brain electrical activity consistent with a reduction in anxiety. The lesions did not significantly decrease hypothalamic-pituitary-adrenal (HPA) activity or cerebrospinal fluid (CSF) concentrations of corticotrophin-releasing factor (CRF). CONCLUSIONS: These findings demonstrate a role for the OFC in mediating anxious temperament and fear-related responses in adolescent primates. Because of the similarities between rhesus monkey threat-induced freezing and childhood behavioral inhibition, these findings are relevant to understanding mechanisms underlying anxious temperament in humans.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Frontal Lobe/physiology , Temperament/physiology , Adrenocorticotropic Hormone/blood , Animals , Anxiety/etiology , Anxiety/pathology , Corticotropin-Releasing Hormone/metabolism , Electroencephalography/methods , Food Preferences/physiology , Freezing Reaction, Cataleptic/physiology , Frontal Lobe/injuries , Hydrocortisone/blood , Macaca mulatta , Naphthalenes , Oxepins , Radioimmunoassay/methods , Reaction Time/physiology
16.
Brain Behav Immun ; 20(6): 564-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16574374

ABSTRACT

Stress impairs healing and in part this effect is thought to be mediated by glucocorticoids. However, the brain systems that underlie the effects of stress on healing remain to be determined. Since the central nucleus of the amygdala (CeA) plays a role in mediating an individual's behavioral and physiological reactivity to stress, we investigated, in rhesus monkeys, whether selective lesions of the CeA altered the gene expression of chemokines (IL-8 and MIP-1alpha) that are associated with early dermal healing. We used rhesus monkeys because they provide an excellent animal model to investigate brain mechanisms relevant to human stress, anxiety, and psychopathology. Hypothalamic-pituitary-adrenal (HPA) activity was assessed in the monkeys prior to the wound healing experiment demonstrating that the CeA lesions reduce HPA activity. In the healing experiment, stress decreased IL-8 and MIP-1alpha gene expression in both CeA lesioned and non-lesioned animals. Conversely, the CeA lesions increased the tissue expression of IL-8 and MIP-1alpha mRNA prior to and after stress exposure. These results demonstrate that in primates the CeA is a key brain region involved in the regulation of processes associated with wound healing. Because of brain and behavioral similarities between rhesus monkeys and humans, these results are particularly relevant to understanding brain mechanisms that influence healing in humans.


Subject(s)
Amygdala/physiology , Interleukin-8/metabolism , Macrophage Inflammatory Proteins/metabolism , Stress, Psychological/immunology , Wound Healing/immunology , Adrenocorticotropic Hormone/blood , Amygdala/immunology , Analysis of Variance , Animals , Chemokine CCL3 , Chemokine CCL4 , Gene Expression Regulation , Hydrocortisone/blood , Interleukin-8/genetics , Macaca mulatta , Macrophage Inflammatory Proteins/genetics , Male , Neuroimmunomodulation , RNA, Messenger/analysis , Skin/immunology , Skin/injuries , Skin/metabolism , Stress, Psychological/metabolism
17.
Biol Psychiatry ; 58(10): 796-804, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16043132

ABSTRACT

BACKGROUND: A key to successful adaptation is the ability to regulate emotional responses in relation to changing environmental demands or contexts. METHODS: High-resolution PET 18fluoro-deoxyglucose (FDG) scanning in rhesus monkeys was performed during two contexts (alone, and human intruder with no eye contact) during which the duration of anxiety related freezing behavior was assessed. Correlations between individual differences in freezing duration and brain activity were performed for each of the two conditions, as well as for the contextual regulation between the two conditions. RESULTS: In both conditions, activity in the basal forebrain, including the bed nucleus of the stria terminalis and the nucleus accumbens were correlated with individual differences in freezing duration. In contrast, individual differences in the ability to regulate freezing behavior between contexts were correlated with activity in the dorsal anterior cingulate cortex, the thalamus and the dorsal raphe nucleus. CONCLUSIONS: These findings demonstrate differences in the neural circuitry mediating the expression compared to the contextual regulation of freezing behavior. These findings are relevant since altered regulatory processes may underlie anxiety disorders.


Subject(s)
Anxiety/psychology , Brain/physiology , Emotions/physiology , Freezing Reaction, Cataleptic/physiology , Amygdala/physiology , Animals , Anxiety/diagnosis , Anxiety/diagnostic imaging , Anxiety Disorders/diagnosis , Anxiety Disorders/psychology , Brain/diagnostic imaging , Brain Mapping , Conditioning, Psychological/physiology , Disease Models, Animal , Fluorodeoxyglucose F18 , Humans , Macaca mulatta , Magnetic Resonance Imaging , Male , Models, Animal , Neural Pathways/physiology , Positron-Emission Tomography , Prefrontal Cortex/physiology , Social Environment
18.
Proc Natl Acad Sci U S A ; 102(11): 4176-9, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15753316

ABSTRACT

In primates, during times of need, calling for help is a universal experience. Calling for help recruits social support and promotes survival. However, calling for help also can attract predators, and it is adaptive to inhibit calls for help when a potential threat is perceived. Based on this, we hypothesized that individual differences in calling for help would be related to the activity of brain systems that mediate goal-directed behavior and the detection of threat. By using high-resolution positron emission tomography in rhesus monkeys undergoing social separation, we demonstrate that increased [18F]-fluoro-2-deoxy-D-glucose uptake in the right dorsolateral prefrontal cortex and decreased uptake in the amygdala independently predict individual differences in calling for help. When taken together, these two regions account for 76% of the variance in calling for help. This result suggests that the drive for affiliation and the perception of threat determine the intensity of an individual's behavior during separation. These findings in monkeys are relevant to humans and provide a conceptual neural framework to understand individual differences in how primates behave when in need of social support.


Subject(s)
Animal Communication , Behavior, Animal/physiology , Brain/physiology , Perception/physiology , Animals , Macaca mulatta , Male , Positron-Emission Tomography
19.
J Neurosci ; 24(24): 5506-15, 2004 Jun 16.
Article in English | MEDLINE | ID: mdl-15201323

ABSTRACT

Numerous studies demonstrate that the rhesus monkey is an excellent species with which to investigate mechanisms underlying human emotion and psychopathology. To examine the role of the central nucleus of the amygdala (CeA) in mediating the behavioral and physiological responses associated with fear and anxiety, we used rhesus monkeys to assess the effects of excitotoxic lesions of the CeA. Behavioral and physiological responses of nine monkeys with bilateral CeA destruction (ranging from 46 to 98%) were compared with five animals with asymmetric lesions (42-86.5% destruction on the most affected side) and with 16 unoperated controls. Results suggest that similar to rodent species, the primate CeA plays a role in mediating fear- and anxiety-related behavioral and endocrine responses. Compared with controls and the asymmetric-lesion group, bilaterally lesioned monkeys displayed significantly less fear-related behavior when exposed to a snake and less freezing behavior when confronted by a human intruder. In addition, bilaterally lesioned monkeys had decreased levels of CSF corticotrophin-releasing factor (CRF), and both lesioned groups had decreased plasma ACTH concentrations. In contrast to these findings, patterns of asymmetric frontal brain electrical activity, as assessed by regional scalp EEG, did not significantly differ between control and lesioned monkeys. These findings suggest that in primates, the CeA is involved in mediating fear- and anxiety-related behavioral and pituitary-adrenal responses as well as in modulating brain CRF activity.


Subject(s)
Amygdala/physiology , Anxiety/physiopathology , Fear/physiology , Animals , Corticotropin-Releasing Hormone/cerebrospinal fluid , Electroencephalography , Humans , Macaca mulatta , Pituitary-Adrenal System/physiology
20.
Emotion ; 3(1): 3-11, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12899313

ABSTRACT

The authors demonstrated individual differences in inhibited behavior and withdrawal responses of laboratory-born rhesus monkeys when initially exposed to a snake. Most monkeys displayed a small significant increase in their behavioral inhibition in the presence of a snake. A few monkeys had marked responses, and some actively withdrew. Although the responses of the most extreme laboratory-born monkeys were comparable to feral-born monkeys, the responses of the laboratory-born monkeys rapidly habituated. The individual differences in the responses of naïve monkeys likely reflect a continuum from orienting to wariness to fear. A neurobiological model is presented that addresses potential mechanisms underlying these individual differences, their relation to fear, and how they may predispose to phobia development.


Subject(s)
Arousal , Fear , Individuality , Macaca mulatta/psychology , Snakes , Animals , Attention , Escape Reaction , Female , Habituation, Psychophysiologic , Male , Phobic Disorders/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...