Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 69: 113-123, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29126979

RESUMO

OBJECTIVE: Mounting evidence indicates that stress influences the experience of pain. Exposure to psychosocial stress disrupts bi-directional communication pathways between the central nervous system and peripheral immune system, and can exacerbate the frequency and severity of pain experienced by stressed subjects. Repeated social defeat (RSD) is a murine model of psychosocial stress that recapitulates the immune and behavioral responses to stress observed in humans, including activation of stress-reactive neurocircuitry and increased pro-inflammatory cytokine production. It is unclear, however, how these stress-induced neuroimmune responses contribute to increased pain sensitivity in mice exposed to RSD. Here we used a technique of regional analgesia with local anesthetics in mice to block the development of mechanical allodynia during RSD. We next investigated the degree to which pain blockade altered stress-induced neuroimmune activation and depressive-like behavior. METHODS: Following development of a mouse model of regional analgesia with discrete sensory blockade over the dorsal-caudal aspect of the spine, C57BL/6 mice were divided into experimental groups and treated with Ropivacaine (0.08%), Liposomal Bupivacaine (0.08%), or Vehicle (0.9% NaCl) prior to exposure to stress. This specific region was selected for analgesia because it is the most frequent location for aggression-associated pain due to biting during RSD. Mechanical allodynia was assessed 12 h after the first, third, and sixth day of RSD after resolution of the sensory blockade. In a separate experiment, social avoidance behavior was determined after the sixth day of RSD. Blood, bone marrow, brain, and spinal cord were collected for immunological analyses after the last day of RSD in both experiments following behavioral assessments. RESULTS: RSD increased mechanical allodynia in an exposure-dependent manner that persisted for at least one week following cessation of the stressor. Mice treated with either Ropivacaine or Liposomal Bupivacaine did not develop mechanical allodynia following exposure to stress, but did develop social avoidance behavior. Neither drug affected stress-induced activation of monocytes in the bone marrow, blood, or brain. Neuroinflammatory responses developed in all treatment groups, as evidenced by elevated IL-1ß mRNA levels in the brain and spinal cord after RSD. CONCLUSIONS: In this study, psychosocial stress was associated with increased pain sensitivity in mice. Development of mechanical allodynia with RSD was blocked by regional analgesia with local anesthetics, Ropivacaine or Liposomal Bupivacaine. Despite blocking mechanical allodynia, these anesthetic interventions did not prevent neuroimmune activation or social avoidance associated with RSD. These data suggest that stress-induced neuroinflammatory changes are not associated with increased sensitivity to pain following RSD. Thus, blocking peripheral nociception was effective in inhibiting enhanced pain signaling without altering stress-induced immune or behavioral responses.


Assuntos
Anestésicos Locais/uso terapêutico , Bupivacaína/uso terapêutico , Limiar da Dor/efeitos dos fármacos , Dor/prevenção & controle , Ropivacaina/uso terapêutico , Comportamento Social , Estresse Psicológico/complicações , Anestésicos Locais/farmacologia , Animais , Comportamento Animal/fisiologia , Bupivacaína/farmacologia , Modelos Animais de Doenças , Camundongos , Dor/etiologia , Dor/imunologia , Medição da Dor , Ropivacaina/farmacologia , Estresse Psicológico/imunologia
2.
Biol Psychiatry ; 79(10): 803-813, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26281717

RESUMO

BACKGROUND: Neuroinflammatory signaling may contribute to the pathophysiology of chronic anxiety disorders. Previous work showed that repeated social defeat (RSD) in mice promoted stress-sensitization that was characterized by the recurrence of anxiety following subthreshold stress 24 days after RSD. Furthermore, splenectomy following RSD prevented the recurrence of anxiety in stress-sensitized mice. We hypothesize that the spleen of RSD-exposed mice became a reservoir of primed monocytes that were released following neuroendocrine activation by subthreshold stress. METHODS: Mice were subjected to subthreshold stress (i.e., single cycle of social defeat) 24 days after RSD, and immune and behavioral measures were taken. RESULTS: Subthreshold stress 24 days after RSD re-established anxiety-like behavior that was associated with egress of Ly6C(hi) monocytes from the spleen. Moreover, splenectomy before RSD blocked monocyte trafficking to the brain and prevented anxiety-like behavior following subthreshold stress. Splenectomy, however, had no effect on monocyte accumulation or anxiety when determined 14 hours after RSD. In addition, splenocytes cultured 24 days after RSD exhibited a primed inflammatory phenotype. Peripheral sympathetic inhibition before subthreshold stress blocked monocyte trafficking from the spleen to the brain and prevented the re-establishment of anxiety in RSD-sensitized mice. Last, ß-adrenergic antagonism also prevented splenic monocyte egress after acute stress. CONCLUSIONS: The spleen served as a unique reservoir of primed monocytes that were readily released following sympathetic activation by subthreshold stress that promoted the re-establishment of anxiety. Collectively, the long-term storage of primed monocytes in the spleen may have a profound influence on recurring anxiety disorders.


Assuntos
Ansiedade/fisiopatologia , Monócitos/fisiologia , Baço/fisiopatologia , Estresse Psicológico/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Animais , Ansiedade/etiologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Estudos de Coortes , Modelos Animais de Doenças , Dominação-Subordinação , Guanetidina/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Baço/efeitos dos fármacos , Esplenectomia , Estresse Psicológico/complicações , Sistema Nervoso Simpático/efeitos dos fármacos , Simpatolíticos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA