Differences in behavioral roles, anatomical connectivity and gene expression patterns in the dorsal, intermediate and ventral regions of the hippocampus are well characterized. Relatively fewer studies have, however, focused on comparing the physiological properties of neurons located at different dorsoventral extents of the hippocampus. Recently we reported that dorsal CA1 neurons are less excitable than ventral neurons. There is little or no information for how neurons in the intermediate hippocampus compare to those from the dorsal and ventral ends. Also, it is not known whether the transition of properties along the dorsoventral axis is gradual or segmented. In this study, we developed a statistical model to predict the dorsoventral position of transverse hippocampal slices. Using current clamp recordings combined with this model, we found that CA1 neurons in dorsal, intermediate and ventral hippocampus have distinct electrophysiological and morphological properties and that the transition in most (but not all) of these properties from the ventral to dorsal end is gradual. Using linear and segmented regression analyses, we found that input resistance and resting membrane potential changed linearly along the V–D axis. Interestingly, the transition in resonance frequency, rebound slope, dendritic branching in stratum radiatum and action potential properties was segmented along the V–D axis. Together, the findings from this study highlight the heterogeneity in CA1 neuronal properties along the entire longitudinal axis of hippocampus.
The development of fetal brain is influenced by nutrients such as docosahexaenoic acid (DHA, 22:6) and choline. Phosphatidylethanolamine-N-methyltransferase (PEMT) catalyzes the biosynthesis of phosphatidylcholine from phosphatidylethanolamine enriched in DHA and many humans have functional genetic polymorphisms in the PEMT gene. Previously, it was reported that Pemt ؊/؊ mice have altered hippocampal development. The present study explores whether abnormal phosphatidylcholine biosynthesis causes altered incorporation of DHA into membranes, thereby influencing brain development, and determines whether supplemental dietary DHA can reverse some of these changes. Pregnant C57BL/6 wild type (WT) and Pemt ؊/؊ mice were fed a control diet, or a diet supplemented with 3 g/kg of DHA, from gestational day 11 to 17. Brains from embryonic day 17 fetuses derived from Pemt ؊/؊ dams fed the control diet had 25-50% less phospholipid-DHA as compared with WT (p < 0.05). Also, they had 60% more neural progenitor cell proliferation (p < 0.05), 60% more neuronal apoptosis (p < 0.01), and 30% less calretinin expression (p < 0.05; a marker of neuronal differentiation) in the hippocampus compared with WT. The DHA-supplemented diet increased fetal brain Pemt ؊/؊ phospholipid-DHA to WT levels, and abrogated the neural progenitor cell proliferation and apoptosis differences. Although this diet did not change proliferation in the WT group, it halved the rate of apoptosis (p < 0.05). In both genotypes, the DHA-supplemented diet increased calretinin expression 2-fold (p < 0.05). These results suggest that the changes in hippocampal development in the Pemt ؊/؊ mouse could be mediated by altered DHA incorporation into membrane phospholipids, and that maternal dietary DHA can influence fetal brain development.
Systemic inflammation associated with a wide range of clinical disorders elicits neuroinflammation in the CNS, exacerbating neurodegeneration or traumatic/ischemic injuries. Neuroinflammation also impairs important forms of neuroplasticity associated with cognitive and motor learning. Several mechanisms propagate systemic inflammation into the CNS, although important details are not yet understood. Reports suggest propagation via vagal pro—inflammatory sensory afferent neurons, and/or cytokines crossing the blood-brain barrier. Microglia are the resident CNS immune cells, and are key contributors to CNS neuroinflammation, producing cytokines, chemokines, NO and ROS. Important contributions are also known from other CNS cells ( e.g. astrocytes, pericytes and neurons). Here, we investigated CNS responses to systemic inflammation induced by systemic LPS injection (1mg/kg, i.p.) in 3 months old Sprague-Dawley rats to test an early stage in the neuroinflammatory process. Three hours post-LPS injection, rats were perfused with PBS followed by microglia isolation from cortex and cervical spinal cord via immunomagnetic separation; the microglia-free fraction (neurons, astrocytes, pericytes, and other cells) was also collected for analysis. The efficacy of microglial isolation was verified by flow cytometry, and both microglial and non-microglial mRNAs were analyzed via qRT-PCR. CD200 and CX3CL1 (fractalkine) are two proteins expressed exclusively by neurons that mediate neuron-microglial communication by interacting with microglial receptors. Post-LPS, both CD200 and CX3CL1 increased 2.5 and 8 fold, respectively. Colony stimulating factor 1 (CSF-1), a protein produced by astrocytes and oligodendrocytes that is crucial for microglial survival, increased 5-fold. Surprisingly, inflammatory cytokines IL6, IL1β, CCL2, iNOS and TNFα were significantly elevated in non-microglial cells, but only IL6 and iNOS were increased in microglia 3 hours post-LPS. Thus, non-microglial cell responses to systemic inflammation appear to occur prior to full microglial activation. We hypothesize that peripheral pro-inflammatory signals propagate into CNS via pericytes and/or astrocytes that are in close contact with blood vessels. Further studies are needed to identify specific cellular sources of pro-inflammatory cytokines. Since CD200 prevents excessive inflammation in peripheral tissues, we also hypothesize that neuronal CD200 upregulation limits microglial immune responses, preventing inappropriate over-activation. Supported by: R01 HL149800 & HL147554 This is the full abstract presented at the American Physiology Summit 2023 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
Dietary docosahexaenoic acid modulates hippocampal development in the mousePhosphatidyl‐N‐methyl transferase (PEMT) catalyzes the biosynthesis of phosphatidyl choline (PtdCho), and prefers incorporation of docosahexaenoic acid (DHA) into PtdCho. We previously reported that the PEMT knock out mouse has altered hippocampal development. The present study explores if changes in membrane DHA species of PtdCho mediate the observed effects on brain development. Timed pregnant CBL57 wild type (WT) mice and PEMT knock‐out (KO) mice were kept on control diet. A subgroup of dams, were given DHA, from embryonic day E11 to E17.Brains of fetuses collected on E17 were processed for immunohistochemistry. Results show increased neural progenitor cell proliferation in the ventricular lining of the developing hippocampus in fetuses from WT dams (92± 29 mitotic cells in DHA treated vs 33±11on control diet, p<0.05), but no change seen in calretenin expressing neurons in the dentate gyrus (DG). In addition, while fetuses of KO dams versus WT dams on control diet showed increased cell proliferation (144± 21 mitotic cells in KO vs 33±11 in WT, p<0.001), fetuses of KO dams on DHA diet did not.In addition, fetuses from KO dams on DHA diet had increased calretenin neurons in DG (3± 0.4 mean optical density in DHA treated vs −0.1 ± 0.2 on control diet, p<0.05) while fetuses from WT dams on DHA diet did not. These data suggest that, maternal dietary DHA, and PEMT‐mediated PtdCho‐DHA in brain can alter hippocampal development.
Therapeutic acute intermittent hypoxia (tAIH) is emerging as a simple and effective means of improving motor function after spinal cord injury (SCI) and ALS. For example, daily AIH applied one-week after C2 spinal hemisection (C2Hx) improves breathing in rats. Unfortunately, studies using the same protocol with chronic cSCI (>8 weeks) report less robust recovery, except when the rats are pretreated with an adenosine 2A receptor antagonist. Pro-inflammatory conditions, such as those experienced after SCI, increase CNS extracellular adenosine concentrations. Guided by increased understanding of adenosinergic signaling mechanisms constraining AIH-induced phrenic motor plasticity, we aimed to determine real-time in vivo dynamics of extracellular adenosine in response to hypoxia in the ventral cervical spinal cord in intact and spinally injured rats. We used enzyme-immobilized micro-biosensors for real time enzymatic detection of extracellular adenosine in the mid-cervical spinal cord of intact rats and rats with chronic C2Hx (>8 weeks post-injury) during 5-min 13% O2, 1-min 9% O2 and 1-min 9% O2 + 4% CO2. Extracellular adenosine exhibits rapid kinetics with a time-constant of seconds, regardless of condition. Rats with chronic C2Hx were classified post hoc into 2 groups: normotensive (systolic blood pressure > 120 mmHg) and hypotensive (systolic blood pressure < 120 mmHg). Normotensive C2Hx rats had baseline adenosine levels comparable to intact rats; hypoxia-induced increases in adenosine were slightly reduced versus intact rats. On the other hand, hypotensive C2Hx rats had elevated baseline adenosine levels and almost completely diminished hypoxia-evoked adenosine increases. Hypercapnic hypoxia had minimal impact on adenosine levels during exposures, but lowered adenosine following each hypercapnic hypoxic episode. These results suggest that changes in cardiovascular hemodynamics in chronic SCI rats can alter basal tissue adenosine levels, possibly undermining the therapeutic efficacy of tAIH in the condition of chronic SCI. Further, hypercapnic hypoxia appears to minimize inhibitory effects of adenosine on tAIH-induced phrenic/diaphragm motor plasticity. Supported by: NIH HL147554, HL148030 & OT2OD023854 and the University of Florida McKnight Brain Institute This is the full abstract presented at the American Physiology Summit 2023 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.