Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-38014350

RESUMO

Scientific evidence underscores the influence of biological sex on the interplay between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress jointly contribute to metabolic dysregulation in both males and females. To address this gap, our study aimed to investigate the combined effects of a high-fat diet (HFD) and repeated footshock stress on fear-related behaviors and metabolic outcomes in male and female mice. Using a robust rodent model that recapitulates key aspects of post-traumatic stress disorder (PTSD), we subjected mice to footshock stressor followed by weekly reminder footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. Our findings revealed that HFD impaired fear memory extinction in male mice that received initial stressor but not in female mice. Blood glucose levels were influenced by both diet and sex, with HFD-fed female mice displaying elevated levels that returned to baseline in the absence of stress, a pattern not observed in male mice. Male mice on HFD exhibited higher energy expenditure, while HFD-fed female mice showed a decreased respiratory exchange ratio (RER). Sex-specific alterations in pro-inflammatory markers and abundance of hematopoietic stem cells were observed in chronically stressed mice on an HFD in different peripheral tissues, indicating the manifestation of distinct comorbid disorders. Single-nuclei RNA sequencing of the ventromedial hypothalamus from stressed mice on an HFD provided insights into sex-specific glial cell activation and cell-type-specific transcriptomic changes. In conclusion, our study offers a comprehensive understanding of the intricate interactions between stress, diet, sex, and various physiological and behavioral outcomes, shedding light on a potential brain region coordinating these interactions.

2.
Nature ; 620(7972): 192-199, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37495690

RESUMO

Sympathetic activation during cold exposure increases adipocyte thermogenesis via the expression of mitochondrial protein uncoupling protein 1 (UCP1)1. The propensity of adipocytes to express UCP1 is under a critical influence of the adipose microenvironment and varies between sexes and among various fat depots2-7. Here we report that mammary gland ductal epithelial cells in the adipose niche regulate cold-induced adipocyte UCP1 expression in female mouse subcutaneous white adipose tissue (scWAT). Single-cell RNA sequencing shows that glandular luminal epithelium subtypes express transcripts that encode secretory factors controlling adipocyte UCP1 expression under cold conditions. We term these luminal epithelium secretory factors 'mammokines'. Using 3D visualization of whole-tissue immunofluorescence, we reveal sympathetic nerve-ductal contact points. We show that mammary ducts activated by sympathetic nerves limit adipocyte UCP1 expression via the mammokine lipocalin 2. In vivo and ex vivo ablation of mammary duct epithelium enhance the cold-induced adipocyte thermogenic gene programme in scWAT. Since the mammary duct network extends throughout most of the scWAT in female mice, females show markedly less scWAT UCP1 expression, fat oxidation, energy expenditure and subcutaneous fat mass loss compared with male mice, implicating sex-specific roles of mammokines in adipose thermogenesis. These results reveal a role of sympathetic nerve-activated glandular epithelium in adipocyte UCP1 expression and suggest that mammary duct luminal epithelium has an important role in controlling glandular adiposity.


Assuntos
Adipócitos , Tecido Adiposo Branco , Epitélio , Glândulas Mamárias Animais , Termogênese , Animais , Feminino , Masculino , Camundongos , Adipócitos/metabolismo , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/metabolismo , Epitélio/inervação , Epitélio/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/inervação , Glândulas Mamárias Animais/fisiologia , Temperatura Baixa , Sistema Nervoso Simpático/fisiologia , Metabolismo Energético , Oxirredução , Caracteres Sexuais
3.
Neurobiol Learn Mem ; 203: 107792, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37369343

RESUMO

Pituitary adenylate cyclase-activating peptide (PACAP) is a highly conserved and widely expressed neuropeptide that has emerged as a key regulator of multiple neural and behavioral processes. PACAP systems, including the various PACAP receptor subtypes, have been implicated in neural circuits of learning and memory, stress, emotion, feeding, and pain. Dysregulation within these PACAP systems may play key roles in the etiology of pathological states associated with these circuits, and PACAP function has been implicated in stress-related psychopathology, feeding and metabolic disorders, and migraine. Accordingly, central PACAP systems may represent important therapeutic targets; however, substantial heterogeneity in PACAP systems related to the distribution of multiple PACAP isoforms across multiple brain regions, as well as multiple receptor subtypes with several isoforms, signaling pathways, and brain distributions, provides both challenges and opportunities for the development of new clinically-relevant strategies to target the PACAP system in health and disease. Here we review the heterogeneity of central PACAP systems, as well as the data implicating PACAP systems in clinically-relevant behavioral processes, with a particular focus on the considerable evidence implicating a role of PACAP in stress responding and learning and memory. We also review data suggesting that there are sex differences in PACAP function and its interactions with sex hormones. Finally, we discuss both the challenges and promise of harnessing the PACAP system in the development of new therapeutic avenues and highlight PACAP systems for their critical role in health and disease.


Assuntos
Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Feminino , Humanos , Masculino , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Aprendizagem , Emoções , Transdução de Sinais/fisiologia
4.
Exp Neurol ; 362: 114339, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36717013

RESUMO

Large scale studies in populations of European and Han Chinese ancestry found a series of rare gain-of-function microduplications in VIPR2, encoding VPAC2, a receptor that binds vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide with high affinity, that were associated with an up to 13-fold increased risk for schizophrenia. To address how VPAC2 receptor overactivity might affect brain development, we used a well-characterized Nestin-Cre mouse strain and a knock-in approach to overexpress human VPAC2 in the central nervous system. Mice that overexpressed VPAC2 were found to exhibit a significant reduction in brain weight. Magnetic resonance imaging analysis confirmed a decrease in brain size, a specific reduction in the hippocampus grey matter volume and a paradoxical increase in whole-brain white matter volume. Sex-specific changes in behavior such as impaired prepulse inhibition and contextual fear memory were observed in VPAC2 overexpressing mice. The data indicate that the VPAC2 receptor may play a critical role in brain morphogenesis and suggest that overactive VPAC2 signaling during development plays a mechanistic role in some forms of schizophrenia.


Assuntos
Receptores Tipo II de Peptídeo Intestinal Vasoativo , Substância Branca , Masculino , Humanos , Feminino , Camundongos , Animais , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Substância Branca/metabolismo , Peptídeo Intestinal Vasoativo/química , Peptídeo Intestinal Vasoativo/metabolismo , Peptídeo Intestinal Vasoativo/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Inibição Pré-Pulso
5.
Neurosci Biobehav Rev ; 142: 104884, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36174795

RESUMO

Extreme stress can cause long-lasting changes in affective behavior manifesting in conditions such as post-traumatic stress disorder (PTSD). Understanding the biological mechanisms that govern trauma-induced behavioral dysregulation requires reliable and rigorous pre-clinical models that recapitulate multiple facets of this complex disease. For decades, Pavlovian fear conditioning has been a dominant paradigm for studying the effects of trauma through an associative learning framework. However, severe stress also causes long-lasting nonassociative fear sensitization, which is often overlooked in Pavlovian fear conditioning studies. This paper synthesizes recent research on the stress-enhanced fear learning (SEFL) paradigm, a valuable rodent model that can dissociate associative and nonassociative effects of stress. We discuss evidence that the SEFL paradigm produces nonassociative fear sensitization that is distinguishable from Pavlovian fear conditioning. We also discuss key biological variables, such as age and sex, neural circuit mechanisms, and crucial gaps in knowledge. We argue that nonassociative fear sensitization deserves more attention within current PTSD models and that SEFL provides a valuable complement to Pavlovian conditioning research on trauma-related pathology.


Assuntos
Medo , Transtornos de Estresse Pós-Traumáticos , Animais , Medo/fisiologia , Aprendizagem/fisiologia , Condicionamento Clássico , Transtornos de Estresse Pós-Traumáticos/psicologia , Roedores , Extinção Psicológica/fisiologia
6.
J Neurosci ; 41(15): 3446-3461, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33637560

RESUMO

Trauma can cause dysfunctional fear regulation leading some people to develop disorders, such as post-traumatic stress disorder (PTSD). The amygdala regulates fear, whereas PACAP (pituitary adenylate activating peptide) and PAC1 receptors are linked to PTSD symptom severity at genetic/epigenetic levels, with a strong link in females with PTSD. We discovered a PACAPergic projection from the basomedial amygdala (BMA) to the medial intercalated cells (mICCs) in adult mice. In vivo optogenetic stimulation of this pathway increased CFOS expression in mICCs, decreased fear recall, and increased fear extinction. Selective deletion of PAC1 receptors from the mICCs in females reduced fear acquisition, but enhanced fear generalization and reduced fear extinction in males. Optogenetic stimulation of the BMA-mICC PACAPergic pathway produced EPSCs in mICC neurons, which were enhanced by the PAC1 receptor antagonist, PACAP 6-38. Our findings show that mICCs modulate contextual fear in a dynamic and sex-dependent manner via a microcircuit containing the BMA and mICCs, and in a manner that was dependent on behavioral state.SIGNIFICANCE STATEMENT Traumatic stress can affect different aspects of fear behaviors, including fear learning, generalization of learned fear to novel contexts, how the fear of the original context is recalled, and how fear is reduced over time. While the amygdala has been studied for its role in regulation of different aspects of fear, the molecular circuitry of this structure is quite complex. In addition, aspects of fear can be modulated differently in males and females. Our findings show that a specific circuitry containing the neuropeptide PACAP and its receptor, PAC1, regulates various aspects of fear, including acquisition, generalization, recall, and extinction in a sexually dimorphic manner, characterizing a novel pathway that modulates traumatic fear.


Assuntos
Tonsila do Cerebelo/fisiologia , Medo , Neurônios/fisiologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Tonsila do Cerebelo/citologia , Animais , Potenciais Pós-Sinápticos Excitadores , Extinção Psicológica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Optogenética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Fatores Sexuais
7.
Anal Bioanal Chem ; 412(23): 5671-5681, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32627085

RESUMO

The direct detection of dopamine (DA) in human body fluids is a great challenge for medical diagnostics of neurological disorders like Parkinson's disease, Alzheimer's disease, senile dementia, and schizophrenia. In this work, a simple and turn off luminescence sensing of DA based on bovine serum albumin (BSA)-capped manganese-doped zinc sulphide quantum dots (Mn:ZnS/BSA QDs) is developed. The Mn:ZnS/BSA QDs were synthesized by a chemical co-precipitation method. Due to the special interaction of DA with BSA and metal ions, Mn:ZnS/BSA QDs can serve as an effective sensing platform for DA. The luminescence of Mn:ZnS/BSA QDs decreased linearly with increasing concentration of DA in the range from 6.6 to 50.6 nM. The limit of detection is 2.02 nM. The driving force for the luminescence quenching is partly provided by ground-state complex formation of QDs with DA. The photo-induced electron transfer from the conduction band of QDs to oxidized dopamine (quinone) also favors quenching. The Mn:ZnS/BSA QDs are barely interfered with by other competing biomolecules except catecholamine neurotransmitter like epinephrine. Moreover, this method is used in the analysis of DA-spiked human serum and human urine samples and good recovery percentages are found. To assess the utility of the developed sensor, paper strip assay was also successfully conducted. Graphical abstract.


Assuntos
Dopamina/química , Manganês/química , Pontos Quânticos/química , Soroalbumina Bovina/química , Sulfetos/química , Compostos de Zinco/química , Dopamina/sangue , Dopamina/urina , Humanos , Concentração de Íons de Hidrogênio , Luminescência , Papel , Análise Espectral/métodos
8.
Neuropsychopharmacology ; 45(3): 482-490, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31787748

RESUMO

Despite the large comorbidity between PTSD and opioid use disorders, as well as the common treatment of physical injuries resulting from trauma with opioids, the ability of opioid treatments to subsequently modify PTSD-related behavior has not been well studied. Using the stress-enhanced fear learning (SEFL) model for PTSD, we characterized the impact of chronic opioid regimens on the sensitization of fear learning seen following traumatic stress in mice. We demonstrate for the first time that chronic opioid pretreatment is able to robustly augment associative fear learning. Highlighting aversive learning as the cognitive process mediating this behavioral outcome, these changes were observed after a considerable period of drug cessation, generalized to learning about multiple aversive stimuli, were not due to changes in stimulus sensitivity or basal anxiety, and correlated with a marker of synaptic plasticity within the basolateral amygdala. Additionally, these changes were not observed when opioids were given after the traumatic event. Moreover, we found that neither reducing the frequency of opioid administration nor bidirectional manipulation of acute withdrawal impacted the subsequent enhancement in fear learning seen. Given the fundamental role of associative fear learning in the generation and progression of PTSD, these findings are of direct translational relevance to the comorbidity between opioid dependence and PTSD, and they are also pertinent to the use of opioids for treating pain resulting from traumas involving physical injuries.


Assuntos
Analgésicos Opioides/administração & dosagem , Medo/efeitos dos fármacos , Medo/psicologia , Aprendizagem/efeitos dos fármacos , Morfina/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/psicologia , Transtornos de Estresse Pós-Traumáticos/psicologia , Analgésicos Opioides/efeitos adversos , Animais , Implantes de Medicamento , Medo/fisiologia , Feminino , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/efeitos adversos , Transtornos Relacionados ao Uso de Opioides/complicações , Transtornos de Estresse Pós-Traumáticos/complicações
9.
Elife ; 82019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31644425

RESUMO

Immune cells are vital constituents of the adipose microenvironment that influence both local and systemic lipid metabolism. Mice lacking IL10 have enhanced thermogenesis, but the roles of specific cell types in the metabolic response to IL10 remain to be defined. We demonstrate here that selective loss of IL10 receptor α in adipocytes recapitulates the beneficial effects of global IL10 deletion, and that local crosstalk between IL10-producing immune cells and adipocytes is a determinant of thermogenesis and systemic energy balance. Single Nuclei Adipocyte RNA-sequencing (SNAP-seq) of subcutaneous adipose tissue defined a metabolically-active mature adipocyte subtype characterized by robust expression of genes involved in thermogenesis whose transcriptome was selectively responsive to IL10Rα deletion. Furthermore, single-cell transcriptomic analysis of adipose stromal populations identified lymphocytes as a key source of IL10 production in response to thermogenic stimuli. These findings implicate adaptive immune cell-adipocyte communication in the maintenance of adipose subtype identity and function.


Assuntos
Adipócitos/efeitos dos fármacos , Comunicação Celular , Regulação da Expressão Gênica , Subunidade alfa de Receptor de Interleucina-10/metabolismo , Interleucina-10/metabolismo , Linfócitos/metabolismo , Termogênese , Adipócitos/fisiologia , Animais , Camundongos , Análise de Célula Única , Transcrição Gênica
10.
Cell ; 177(5): 1280-1292.e20, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31031006

RESUMO

Hyperactivity and disturbances of attention are common behavioral disorders whose underlying cellular and neural circuit causes are not understood. We report the discovery that striatal astrocytes drive such phenotypes through a hitherto unknown synaptic mechanism. We found that striatal medium spiny neurons (MSNs) triggered astrocyte signaling via γ-aminobutyric acid B (GABAB) receptors. Selective chemogenetic activation of this pathway in striatal astrocytes in vivo resulted in acute behavioral hyperactivity and disrupted attention. Such responses also resulted in upregulation of the synaptogenic cue thrombospondin-1 (TSP1) in astrocytes, increased excitatory synapses, enhanced corticostriatal synaptic transmission, and increased MSN action potential firing in vivo. All of these changes were reversed by blocking TSP1 effects. Our data identify a form of bidirectional neuron-astrocyte communication and demonstrate that acute reactivation of a single latent astrocyte synaptogenic cue alters striatal circuits controlling behavior, revealing astrocytes and the TSP1 pathway as therapeutic targets in hyperactivity, attention deficit, and related psychiatric disorders.


Assuntos
Astrócitos/metabolismo , Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Comportamento Animal , Comunicação Celular , Neurônios/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Animais , Astrócitos/patologia , Transtorno do Deficit de Atenção com Hiperatividade/genética , Transtorno do Deficit de Atenção com Hiperatividade/patologia , Transtorno do Deficit de Atenção com Hiperatividade/fisiopatologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Sinapses/genética , Trombospondina 1/genética , Trombospondina 1/metabolismo , Ácido gama-Aminobutírico/genética , Ácido gama-Aminobutírico/metabolismo
11.
J Cell Physiol ; 234(5): 7368-7383, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30370526

RESUMO

The current study aimed at developing diverse Trichoderma fusants for fungicides, drought, and salt tolerance with enhanced antagonistic activity against Sclerotium rolfsii Sacc. Trichoderma virens NBAII Tvs12 (mycoparasitic) and Trichoderma koningii MTCC796 (multistress tolerant) were used as parental strains for development of interspecific protoplast fusants. A total of 36 stable fusants were used for mycoparasitism, fungicides, and abiotic stresses (drought and salt) tolerance. The results revealed 20 homozygous progenies showing characteristics of either one parental strain and 14 heterozygous mutants depicting traits of both parental strains. A novel concept of inhibition coefficient was established using growth-related key parameters that represent the pathogen biology and the biocontrol-related biophysics of Trichoderma fusants. The results indicated a differential inhibition coefficient of the test pathogen and the highest (92.88%) inhibition coefficient of S. rolfsii was observed by interstable fusant Fu21. It also grew better under fungicides and abiotic stress (drought and salt) conditions. The molecular characterization and heterozygosity analysis evidenced the highest observed heterozygosity (0.5441) and gene flow (0.3872) in stable heterozygous Fu21. Principal coordinates analysis exhibited 62.7% of total variability. The ecofriendly heterozygous Trichoderma fusant (Fu21) might be useful for biocontrol of stem rot disease under adverse conditions or as a part of integrated disease management.


Assuntos
Basidiomycota/crescimento & desenvolvimento , Mutação , Controle Biológico de Vetores , Doenças das Plantas/prevenção & controle , Trichoderma/genética , Basidiomycota/patogenicidade , Desidratação/genética , Secas , Fungicidas Industriais/farmacologia , Heterozigoto , Homozigoto , Doenças das Plantas/microbiologia , Tolerância ao Sal/genética , Trichoderma/efeitos dos fármacos , Trichoderma/crescimento & desenvolvimento , Trichoderma/ultraestrutura
12.
J Vis Exp ; (140)2018 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-30371665

RESUMO

Fear behaviors are important for survival, but disproportionately high levels of fear can increase the vulnerability for developing psychiatric disorders such as post-traumatic stress disorder (PTSD). To understand the biological mechanisms of fear dysregulation in PTSD, it is important to start with a valid animal model of the disorder. This protocol describes the methodology required to conduct stress-enhanced fear learning (SEFL) experiments, a preclinical model of PTSD, in both rats and mice. SEFL was developed to recapitulate critical aspects of PTSD, including long-term sensitization of fear learning caused by an acute stressor. SEFL uses aspects of Pavlovian fear conditioning but produces a distinct and robust sensitized fear response far greater than normal conditional fear responses. The trauma procedure involves placing a rodent in a conditioning chamber and administering 15 unsignaled shocks randomly distributed over 90 minutes (for rat experiments; for mouse experiments, 10 unsignaled shocks randomly distributed over 60 minutes are used). On day 2, rodents are placed in a novel conditioning context where they receive a single shock; then, on day 3 they are placed back in the same context as on day 2 and tested for changes in freezing levels. Rodents that previously received the trauma display enhanced levels of freezing on the test day compared to those that received no shocks on the first day. Thus, with this model, a single highly stressful experience (the trauma) produces extreme fear of the stimuli associated with the traumatic event.


Assuntos
Medo/psicologia , Aprendizagem/fisiologia , Transtornos de Estresse Pós-Traumáticos/psicologia , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Ratos , Roedores
13.
Anal Chim Acta ; 1031: 152-160, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30119734

RESUMO

Hyperbilirubinemia is the condition when bilirubin exceeds normal concentration in body (19.80 mg/mL in newborns and 1.19 × 10-2 mg/mL in adults). Bilirubin encephalopathy in newborns may cause irreversible neurological disorders. Current methods for detection of bilirubin suffer from compromising accuracy. In the present work, bovine serum albumin stabilized copper nanocluster (BSA-CuNCs) was synthesized via a one pot microwave assisted method as a turn on detector for bilirubin. The synthesized BSA-CuNCs having size less than 4 nm, exhibited bright blue emission at 405 nm. Interestingly, no observable change in fluorescence emission was noticed over a wide pH range (1-11) or at high ionic conditions. However, the interaction of Fe3+ with BSA-CuNCs induces quenching of fluorescence. Moreover, the fluorescence can be regained by the addition of bilirubin over other possible coexisting biomolecules. A good linearity was observed for BSA-CuNCs based turn on probe with a Limit of Detection (LoD) 6.62 nM. Furthermore, real sample analyses were carried out with human serum and urine which showed good recovery percentage.


Assuntos
Bilirrubina/análise , Cobre/química , Compostos Férricos/química , Nanopartículas Metálicas/química , Soroalbumina Bovina/química , Espectrometria de Fluorescência , Bilirrubina/sangue , Bilirrubina/urina , Humanos , Concentração de Íons de Hidrogênio , Limite de Detecção , Concentração Osmolar
14.
Mol Genet Metab ; 124(2): 114-123, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29724658

RESUMO

The transplantation, engraftment, and expansion of primary hepatocytes have the potential to be an effective therapy for metabolic disorders of the liver including those of nitrogen metabolism. To date, such methods for the treatment of urea cycle disorders in murine models has only been minimally explored. Arginase deficiency, an inherited disorder of nitrogen metabolism that presents in the first two years of life, has the potential to be treated by such methods. To explore the potential of this approach, we mated the conditional arginase deficient mouse with a mouse model deficient in fumarylacetoacetate hydrolase (FAH) and with Rag2 and IL2-Rγ mutations to give a selective advantage to transplanted (normal) human hepatocytes. On day -1, a uroplasminogen-expressing adenoviral vector was administered intravenously followed the next day with the transplantation of 1 × 106 human hepatocytes (or vehicle alone) by intrasplenic injection. As the initial number of administered hepatocytes would be too low to prevent hepatotoxicity-induced mortality, NTBC cycling was performed to allow for hepatocyte expansion and repopulation. While all control mice died, all except one human hepatocyte transplanted mice survived. Four months after hepatocyte transplantation, 2 × 1011 genome copies of AAV-TBG-Cre recombinase was administered IV to disrupt endogenous hepatic arginase expression. While all control mice died within the first month, human hepatocyte transplanted mice did well. Ammonia and amino acids, analyzed in both groups before and after disruption of endogenous arginase expression, while well-controlled in the transplanted group, were markedly abnormal in the controls. Ammonium challenging further demonstrated the durability and functionality of the human repopulated liver. In conclusion, these studies demonstrate that human hepatocyte repopulation in the murine liver can result in effective treatment of arginase deficiency.


Assuntos
Arginase/fisiologia , Predisposição Genética para Doença , Hepatócitos/transplante , Hepatopatias/terapia , Doenças Metabólicas/terapia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Hepatócitos/citologia , Humanos , Hepatopatias/enzimologia , Hepatopatias/patologia , Masculino , Doenças Metabólicas/enzimologia , Doenças Metabólicas/patologia , Camundongos , Camundongos Knockout
15.
Neurobiol Learn Mem ; 145: 222-231, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29030297

RESUMO

The structurally related neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) have been implicated in stress regulation and learning and memory. Several bodies of research have shown the impact of the PACAP specific receptor PAC1 on fear memory, but the roles of other PACAP receptors in regulating fear stress responses remain to be elucidated. Here we aimed to investigate the effects of genetic deletion of VIPR2 encoding the VPAC2 receptor, which binds both VIP and PACAP, on fear-related memory and on dendritic morphology in the brain regions of the fear circuitry. Male VPAC2 receptor knockout (VPAC2-KO) and littermate wild-type control mice were subjected to Pavlovian fear conditioning paradigm. VPAC2-KO mice displayed normal acquisition of fear conditioning, contextual and cued fear memory, but impaired extinction of cued fear memory. Morphological analyses revealed reductions in cell body size and total branch number and length of apical and basal dendrites of prelimbic cortex neurons in VPAC2-KO mice. In addition, Sholl analysis indicated that the amount of dendritic material distal to the soma was decreased, while proximal dendritic material was increased. In the infralimbic cortex, the amount of apical dendritic material proximal to the soma was increased in VPAC2-KO mice, while other indices of morphology did not differ. Finally, there were no differences in dendritic morphology in basolateral amygdala neurons between genotypes. These findings suggest that the VPAC2 receptor plays an important role in the fear extinction processes and the regulation of the dendritic morphology in the prelimbic and infralimbic cortices.


Assuntos
Dendritos , Extinção Psicológica/fisiologia , Medo/fisiologia , Córtex Pré-Frontal/fisiologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia , Animais , Condicionamento Clássico , Sinais (Psicologia) , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Córtex Pré-Frontal/citologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética
16.
J Neurosci Res ; 94(12): 1393-1399, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27661774

RESUMO

Fear is an important emotional reaction in response to threatening stimuli and is important for survival. However, when fear occurs in inappropriate circumstances, it can lead to pathological conditions including an increased vulnerability for developing anxiety disorders such as posttraumatic stress disorder (PTSD). Patients with PTSD generalize fear to contexts or to environments that are not associated with the trauma. We sought to explore if increasing the level of dissimilarity relative to the context in which mice learn fear results in changes in the level of fear responding to the new context. We also determined with this procedure if the number of cells expressing the immediate early gene cfos changes with the corresponding level of expressed fear within brain regions known to be important in modulating fear, including the basolateral amygdala (BLA) and hippocampus. Our results indicate that mice that were tested in increasingly different contexts showed significantly different levels of fear responses. Freezing level was higher in the context most similar to the acquisition context than the one that was highly different. The level of cfos within the BLA, but not hippocampus, was also significantly different between the test contexts, with higher levels in the somewhat similar compared with the most different context. Overall, these results highlight the BLA as a critical region in the node of fear circuitry for modulating fear generalization. © 2016 Wiley Periodicals, Inc.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/metabolismo , Medo/psicologia , Generalização Psicológica , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Condicionamento Psicológico , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos de Estresse Pós-Traumáticos/psicologia
17.
J Comp Neurol ; 524(18): 3827-3848, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27197019

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP, gene name Adcyap1) regulates a wide variety of neurological and physiological functions, including metabolism and cognition, and plays roles in of multiple forms of stress. Because of its preferential expression in nerve fibers, it has often been difficult to trace and identify the endogenous sources of the peptide in specific populations of neurons. Here, we introduce a transgenic mouse line that harbors in its genome a bacterial artificial chromosome containing an enhanced green fluorescent protein (EGFP) expression cassette inserted upstream of the PACAP ATG translation initiation codon. Analysis of expression in brain sections of these mice using a GFP antibody reveals EGFP expression in distinct neuronal perikarya and dendritic arbors in several major brain regions previously reported to express PACAP from using a variety of approaches, including radioimmunoassay, in situ hybridization, and immunohistochemistry with and without colchicine. EGFP expression in neuronal perikarya was modulated in a manner similar to PACAP gene expression in motor neurons after peripheral axotomy in the ipsilateral facial motor nucleus in the brainstem, providing an example in which the transgene undergoes proper regulation in vivo. These mice and the high-resolution map obtained are expected to be useful in understanding the anatomical patterns of PACAP expression and its plasticity in the mouse. J. Comp. Neurol. 524:3827-3848, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Camundongos Transgênicos , Modelos Animais , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Animais , Axotomia , Encéfalo/citologia , Encéfalo/metabolismo , Traumatismos do Nervo Facial/metabolismo , Traumatismos do Nervo Facial/patologia , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Masculino , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Medula Espinal/citologia , Medula Espinal/metabolismo
18.
Indian J Pathol Microbiol ; 59(1): 87-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26960646

RESUMO

Primary leiomyomas are rare benign tumors of the lung and only 25 cases have been documented in children, most of which are endobronchial. Leiomyomas are benign smooth muscle neoplasms, usually diagnosed on morphological characteristics. However, immunohistochemistry plays a crucial role in the lineage differentiation when these are encountered at unusual sites or with unexpected morphological features. We report a case of endobronchial tumor of a 13-year-old male child who presented with a dry cough and hemoptysis. A mass lesion in the right main bronchus was detected by bronchoscopy and contrast-enhanced computed tomography. On histopathological examination of the resected specimen, a diagnosis of myxoid leiomyoma was made. To the best of our knowledge, this is the first case of myxoid leiomyoma to be reported in the respiratory tract. This case also demonstrates the need for a high index of suspicion and the role of immunomarkers in the diagnosis of such challenging cases.


Assuntos
Brônquios/patologia , Neoplasias Brônquicas/diagnóstico , Neoplasias Brônquicas/patologia , Leiomioma/diagnóstico , Leiomioma/patologia , Actinas/análise , Adolescente , Biomarcadores Tumorais/análise , Neoplasias Brônquicas/diagnóstico por imagem , Broncoscopia , Desmina/análise , Histocitoquímica , Humanos , Imuno-Histoquímica , Leiomioma/diagnóstico por imagem , Masculino , Microscopia , Tomografia Computadorizada por Raios X
19.
J Neurosci ; 35(42): 14270-85, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26490866

RESUMO

The neurobiology of post-traumatic stress disorder (PTSD) remains unclear. Intense stress promotes PTSD, which has been associated with exaggerated startle and deficient sensorimotor gating. Here, we examined the long-term sequelae of a rodent model of traumatic stress (repeated predator exposure) on amygdala systems that modulate startle and prepulse inhibition (PPI), an operational measure of sensorimotor gating. We show in rodents that repeated psychogenic stress (predator) induces long-lasting sensitization of basolateral amygdala (BLA) noradrenergic (NE) receptors (α1) via a corticotropin-releasing factor receptor 1 (CRF-R1)-dependent mechanism, and that these CRF1 and NE α1 receptors are highly colocalized on presumptive excitatory output projection neurons of the BLA. A profile identical to that seen with predator exposure was produced in nonstressed rats by intra-BLA infusions of CRF (200 ng/0.5 µl), but not by repeated NE infusions (20 µg/0.5 µl). Infusions into the adjacent central nucleus of amygdala had no effect. Importantly, the predator stress- or CRF-induced sensitization of BLA manifested as heightened startle and PPI deficits in response to subsequent subthreshold NE system challenges (with intra-BLA infusions of 0.3 µg/0.5 µl NE), up to 1 month after stress. This profile of effects closely resembles aspects of PTSD. Hence, we reveal a discrete neural pathway mediating the enhancement of NE system function seen in PTSD, and we offer a model for characterizing potential new treatments that may work by modulating this BLA circuitry. SIGNIFICANCE STATEMENT: The present findings reveal a novel and discrete neural substrate that could underlie certain core deficits (startle and prepulse inhibition) that are observed in post-traumatic stress disorder (PTSD). It is shown here that repeated exposure to a rodent model of traumatic stress (predator exposure) produces a long-lasting sensitization of basolateral amygdala noradrenergic substrates [via a corticotropin-releasing factor (CRF)-dependent mechanism] that regulate startle, which is exaggerated in PTSD. Moreover, it is demonstrated that the sensitized noradrenergic receptors colocalize with CRF1 receptors on output projection neurons of the basolateral amygdala. Hence, this stress-induced sensitization of noradrenergic receptors on basolateral nucleus efferents has wide-ranging implications for the numerous deleterious sequelae of trauma exposure that are seen in multiple psychiatric illnesses, including PTSD.


Assuntos
Complexo Nuclear Basolateral da Amígdala/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Norepinefrina/metabolismo , Reflexo de Sobressalto/fisiologia , Transtornos de Estresse Pós-Traumáticos/patologia , Estresse Psicológico/patologia , Estimulação Acústica , Análise de Variância , Animais , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Furões , Glutamato Descarboxilase/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Reflexo de Sobressalto/efeitos dos fármacos , Fatores de Tempo , Ácido gama-Aminobutírico/metabolismo
20.
Psychopharmacology (Berl) ; 220(3): 465-79, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21947334

RESUMO

RATIONALE: Prepulse inhibition (PPI), a preattentional information-filtering mechanism, is disrupted by serotonin (5-HT) or norepinephrine (NE) agonists to model deficits seen in schizophrenia, but whether this effect occurs through interactions between these systems is not known. OBJECTIVES: These studies investigated whether PPI/activity changes induced by agonists of one system were dependent on neurotransmission within the other. METHODS: Male Sprague-Dawley rats received the 5-HT(2) receptor agonist DOI (1-[2,5-dimethoxy-4-iodophenyl]-2-aminopropane) (0, 0.3 mg/kg), with or without antagonists for α1 (prazosin:0, 0.3, or 1 mg/kg) or ß (timolol:0, 3, or 10 mg/kg) receptors or their combination (0 or 0.3 mg/kg prazosin + 3 mg/kg timolol), or the 5-HT(2) antagonist ritanserin (0, 2 mg/kg). Separately, the α1-adrenergic receptor agonist cirazoline (0, 0.68 mg/kg) was given with and without ritanserin (0, 0.5, or 2 mg/kg) or the NE antagonists (0 or 0.3 mg/kg prazosin + 3 mg/kg timolol). Finally, combinations of subthreshold doses of DOI (0, 0.01, 0.025 mg/kg) and cirazoline (0, 0.1, 0.25 mg/kg) were tested for their ability to disrupt PPI, and concomitant administration of all three antagonists (0 vs. 0.3 mg/kg prazosin + 3 mg/kg timolol + 2 mg/kg ritanserin) was assessed for its ability to modify PPI. Locomotion was assessed in an additional set of experiments. RESULTS: Doses/combinations of prazosin and timolol that reversed cirazoline-induced effects did not alter DOI-induced effects, and ritanserin did not affect cirazoline at doses that blocked DOI-mediated effects. Concomitant antagonism of α1 + ß + 5-HT(2) receptors did not modify PPI, nor did combinations of subthreshold doses of cirazoline and DOI. CONCLUSIONS: 5-HT(2) receptors and α1 and ß NE receptors may act through independent mechanisms to modulate sensorimotor gating and locomotor activity.


Assuntos
Atividade Motora/efeitos dos fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/metabolismo , Filtro Sensorial/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores 5-HT2 de Serotonina/efeitos dos fármacos , Receptores 5-HT2 de Serotonina/metabolismo , Reflexo de Sobressalto/efeitos dos fármacos , Esquizofrenia/fisiopatologia , Antagonistas da Serotonina/administração & dosagem , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/administração & dosagem , Agonistas do Receptor de Serotonina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...