Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Neurosci ; 17(1): 97-105, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24292233

ABSTRACT

Tumor necrosis factor α (TNF) is a proinflammatory cytokine with established roles in host defense and immune system organogenesis. We studied TNF function and found a previously unidentified physiological function that extends its effect beyond the host into the developing offspring. A partial or complete maternal TNF deficit, specifically in hematopoietic cells, resulted in reduced milk levels of the chemokines IP-10, MCP-1, MCP-3, MCP-5 and MIP-1ß, which in turn augmented offspring postnatal hippocampal proliferation, leading to improved adult spatial memory in mice. These effects were reproduced by the postpartum administration of a clinically used anti-TNF agent. Chemokines, fed to suckling pups of TNF-deficient mothers, restored both postnatal proliferation and spatial memory to normal levels. Our results identify a TNF-dependent 'lactrocrine' pathway that programs offspring hippocampal development and memory. The level of ambient TNF is known to be downregulated by physical activity, exercise and adaptive stress. We propose that the maternal TNF-milk chemokine pathway evolved to promote offspring adaptation to post-weaning environmental challenges and competition.


Subject(s)
Cytokines/metabolism , Hippocampus/growth & development , Memory/physiology , Milk/chemistry , Tumor Necrosis Factor-alpha/metabolism , Analysis of Variance , Animals , Animals, Newborn , Animals, Suckling , Female , Glial Fibrillary Acidic Protein , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/ultrastructure , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nestin/genetics , Pregnancy , RNA, Messenger , SOXB1 Transcription Factors , Silver Staining , T-Box Domain Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics
2.
Front Psychiatry ; 2: 25, 2011.
Article in English | MEDLINE | ID: mdl-21629836

ABSTRACT

Risk factors for psychiatric disorders have traditionally been classified as genetic or environmental. Risk (candidate) genes, although typically possessing small effects, represent a clear starting point to elucidate downstream cellular/molecular pathways of disease. Environmental effects, especially during development, can also lead to altered behavior and increased risk for disease. An important environmental factor is the mother, demonstrated by the negative effects elicited by maternal gestational stress and altered maternal care. These maternal effects can also have a genetic basis (e.g., maternal genetic variability and mutations). The focus of this review is "maternal genotype effects" that influence the emotional development of the offspring resulting in life-long psychiatric disease-like phenotypes. We have recently found that genetic inactivation of the serotonin 1A receptor (5-HT1AR) and the fmr1 gene (encoding the fragile X mental retardation protein) in mouse dams results in psychiatric disease-like phenotypes in their genetically unaffected offspring. 5-HT1AR deficiency in dams results in anxiety and increased stress responsiveness in their offspring. Offspring of 5-HT1AR deficient dams display altered development of the hippocampus, which could be linked to their anxiety-like phenotype. Maternal inactivation of fmr1, like its inactivation in the offspring, results in a hyperactivity-like condition and is associated with receptor alterations in the striatum. These data indicate a high sensitivity of the offspring to maternal mutations and suggest that maternal genotype effects can increase the impact of genetic risk factors in a population by increasing the risk of the genetically normal offspring as well as by enhancing the effects of offspring mutations.

3.
J Neurosci Methods ; 172(2): 173-7, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18533271

ABSTRACT

The dentate gyrus is one of the few brain regions that show proliferation of neuronal precursors postnatally and in adult life. Proliferation in the dentate gyrus has been shown to be influenced by exercise, stress and drugs such as antidepressants. Traditionally, proliferation studies rely on the time consuming and subjective manual count of labeled cells. Here we adapted the Metamorph software to automatically count cells labeled in the S phase in the developing dentate gyrus of mice. The validity of the computer-assisted method was established by showing an outcome similar to that obtained with the established manual counting procedure. In addition, by using a genetically modified mouse line with increased proliferation, the ability of the computer-assisted method to detect changes in proliferation was demonstrated.


Subject(s)
Cell Proliferation , Hippocampus/cytology , Image Cytometry/methods , Neurons/cytology , Stem Cells/cytology , Aging/genetics , Animals , Animals, Newborn , Bromodeoxyuridine , Cell Count/methods , Computers , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Hippocampus/growth & development , Image Cytometry/instrumentation , Mice , Mice, Knockout , Mice, Neurologic Mutants , Neurons/physiology , Reproducibility of Results , S Phase/physiology , Software , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...