Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jane M. Bell is active.

Publication


Featured researches published by Jane M. Bell.


Journal of Lipid Research | 2006

One generation of n-3 polyunsaturated fatty acid deprivation increases depression and aggression test scores in rats.

James C. DeMar; Kiazong Ma; Jane M. Bell; Miki Igarashi; Deanna Greenstein; Stanley I. Rapoport

Male rat pups at weaning (21 days of age) were subjected to a diet deficient or adequate in n-3 polyunsaturated fatty acids (n-3 PUFAs) for 15 weeks. Performance on tests of locomotor activity, depression, and aggression was measured in that order during the ensuing 3 weeks, after which brain lipid composition was determined. In the n-3 PUFA-deprived rats, compared with n-3 PUFA-adequate rats, docosahexaenoic acid (22:6n-3) in brain phospholipid was reduced by 36% and docosapentaenoic acid (22:5n-6) was elevated by 90%, whereas brain phospholipid concentrations were unchanged. N-3 PUFA-deprived rats had a significantly increased (P = 0.03) score on the Porsolt forced-swim test for depression, and increased blocking time (P = 0.03) and blocking number (P = 0.04) scores (uncorrected for multiple comparisons) on the isolation-induced resident-intruder test for aggression. Large effect sizes (d > 0.8) were found on the depression score and on the blocking time score of the aggression test. Scores on the open-field test for locomotor activity did not differ significantly between groups, and had only small to medium effect sizes. This single-generational n-3 PUFA-deprived rat model, which demonstrated significant changes in brain lipid composition and in test scores for depression and aggression, may be useful for elucidating the contribution of disturbed brain PUFA metabolism to human depression, aggression, and bipolar disorder.


Journal of Neurochemistry | 2004

Half-lives of docosahexaenoic acid in rat brain phospholipids are prolonged by 15 weeks of nutritional deprivation of n-3 polyunsaturated fatty acids

James C. DeMar; Kaizong Ma; Jane M. Bell; Stanley I. Rapoport

Male rat pups (21 days old) were placed on a diet deficient in n‐3 polyunsaturated fatty acids (PUFAs) or on an n‐3 PUFA adequate diet containing α‐linolenic acid (α‐LNA; 18 : 3n‐3). After 15 weeks on a diet, [4,5‐3H]docosahexaenoic acid (DHA; 22 : 6n‐3) was injected into the right lateral cerebral ventricle, and the rats were killed at fixed times over a period of 60 days. Compared with the adequate diet, 15 weeks of n‐3 PUFA deprivation reduced plasma DHA by 89% and brain DHA by 37%; these DHA concentrations did not change thereafter. In the n‐3 PUFA adequate rats, DHA loss half‐lives, calculated by plotting log10 (DHA radioactivity) against time after tracer injection, equaled 33 days in total brain phospholipid, 23 days in phosphatidylcholine, 32 days in phosphatidylethanolamine, 24 days in phosphatidylinositol and 58 days in phosphatidylserine; all had a decay slope significantly greater than 0 (p < 0.05). In the n‐3 PUFA deprived rats, these half‐lives were prolonged twofold or greater, and calculated rates of DHA loss from brain, Jout, were reduced. Mechanisms must exist in the adult rat brain to minimize DHA metabolic loss, and to do so even more effectively in the face of reduced n‐3 PUFA availability for only 15 weeks.


Journal of Neurochemistry | 2005

α-Linolenic acid does not contribute appreciably to docosahexaenoic acid within brain phospholipids of adult rats fed a diet enriched in docosahexaenoic acid

James C. DeMar; Kaizong Ma; Lisa Chang; Jane M. Bell; Stanley I. Rapoport

Adult male unanesthetized rats, reared on a diet enriched in both α‐linolenic acid (α‐LNA) and docosahexaenoic acid (DHA), were infused intravenously for 5 min with [1‐14C]α‐LNA. Timed arterial samples were collected until the animals were killed at 5 min and the brain was removed after microwaving. Plasma and brain lipid concentrations and radioactivities were measured. Within plasma lipids, > 99% of radioactivity was in the form of unchanged [1‐14C]α‐LNA. Eighty‐six per cent of brain radioactivity at 5 min was present as β‐oxidation products, whereas the remainder was mainly in ‘stable’ phospholipid or triglyceride as α‐LNA or DHA. Equations derived from kinetic modeling demonstrated that unesterified unlabeled α‐LNA rapidly enters brain from plasma, but that its incorporation into brain phospholipid and triglyceride, as in the form of synthesized DHA, is ≤ 0.2% of the amount that enters the brain. Thus, in rats fed a diet containing large amounts of both α‐LNA and DHA, the α‐LNA that enters brain from plasma largely undergoes β‐oxidation, and is not an appreciable source of DHA within brain phospholipids.


Neuroscience Letters | 1996

Lithium decreases turnover of arachidonate in several brain phospholipids

Michael C. J. Chang; Eric Grange; Olivier Rabin; Jane M. Bell; David D. Allen; Stanley I. Rapoport

In vivo rates of incorporation and turnover of palmitate and arachidonate in brain phospholipids were measured in awake rats treated chronically with lithium, following intravenous infusion of radiolabeled palmitate and arachidonate, respectively. Chronic lithium, at a brain level considered to be therapeutic in humans, decreased turnover of arachidonate within brain phosphatidylinositol, phosphatidylcholine and phosphatidylethanolamine by up to 80% (P < 0.001). In contrast, lithium had a minimal effect on turnover of palmitate, causing only a 26% reduction in turnover in phosphatidylcholine (P < 0.01). These results suggest that a major therapeutic effect of lithium is to reduce turnover specifically of arachidonate, possibly by inhibiting phospholipase A2 involved in signal transduction. The effect may be secondary to the known action of lithium on the phosphoinositide cycle, by inhibiting the activity of inositol monophosphatase.


Journal of Neurochemistry | 2001

Chronic valproate treatment decreases the in vivo turnover of arachidonic acid in brain phospholipids: a possible common effect of mood stabilizers

Michael C. J. Chang; Miguel A. Contreras; Thad A. Rosenberger; Jyrki Rintala; Jane M. Bell; Stanley I. Rapoport

Both (Li+) and valproic acid (VPA) are effective in treating bipolar disorder, but the pathway by which either works, and whether it is common to both drugs, is not agreed upon. We recently reported, using an in vivo fatty acid model, that Li+ reduces the turnover rate of the second messenger arachidonic acid (AA) by 80% in brain phospholipids of the awake rat, without changing turnover rates of docosahexaenoic or palmitic acid. Reduced AA turnover was accompanied by down‐regulation of gene expression and protein levels of an AA‐specific cytosolic phospholipase A2 (cPLA2). To see if VPA had the same effect on AA turnover, we used our in vivo fatty acid model in rats chronically administered VPA (200 mg/kg, i.p. for 30 days). Like Li+, VPA treatment significantly decreased AA turnover within brain phospholipids (by 28–33%), although it had no effect on cPLA2 protein levels. Thus, both mood stabilizers, Li+ and VPA have a common action in reducing AA turnover in brain phospholipids, albeit by different mechanisms.


Journal of Lipid Research | 2007

Dietary n-3 PUFA deprivation for 15 weeks upregulates elongase and desaturase expression in rat liver but not brain

Miki Igarashi; Kaizong Ma; Lisa Chang; Jane M. Bell; Stanley I. Rapoport

Fifteen weeks of dietary n-3 PUFA deprivation increases coefficients of conversion of circulating α-linolenic acid (α-LNA; 18:3n-3) to docosahexaenoic acid (DHA; 22:6n-3) in rat liver but not brain. To determine whether these increases reflect organ differences in enzymatic activities, we examined brain and liver expression of converting enzymes and of two of their transcription factors, peroxisome proliferator-activated receptor α (PPARα) and sterol-regulatory element binding protein-1 (SREBP-1), in rats fed an n-3 PUFA “adequate” (4.6% α-LNA of total fatty acid, no DHA) or “deficient” (0.2% α-LNA, no DHA) diet for 15 weeks after weaning. In rats fed the deficient compared with the adequate diet, mRNA and activity levels of Δ5 and Δ6 desaturases and elongases 2 and 5 were upregulated in liver but not brain, but liver PPARα and SREBP-1 mRNA levels were unchanged. In rats fed the adequate diet, enzyme activities generally were higher in liver than brain. Thus, differences in conversion enzyme expression explain why the liver has a greater capacity to synthesize DHA from circulating α-LNA than does the brain in animals on an adequate n-3 PUFA diet and why liver synthesis capacity is increased by dietary deprivation. These data suggest that liver n-3 PUFA metabolism determines DHA availability to the brain when DHA is absent from the diet.


Neuropsychopharmacology | 2006

Chronic Lithium Chloride Administration Attenuates Brain NMDA Receptor-Initiated Signaling via Arachidonic Acid in Unanesthetized Rats

Mireille Basselin; Lisa Chang; Jane M. Bell; Stanley I. Rapoport

It has been proposed that lithium is effective in bipolar disorder (BD) by inhibiting glutamatergic neurotransmission, particularly via N-methyl-D-aspartate receptors (NMDARs). To test this hypothesis and to see if the neurotransmission could involve the NMDAR-mediated activation of phospholipase A2 (PLA2), to release arachidonic acid (AA) from membrane phospholipid, we administered subconvulsant doses of NMDA to unanesthetized rats fed a chronic control or LiCl diet. We used quantitative autoradiography following the intravenous injection of radiolabeled AA to measure regional brain incorporation coefficients k* for AA, which reflect receptor-mediated activation of PLA2. In control diet rats, NMDA (25 and 50 mg/kg i.p.) compared with i.p. saline increased k* significantly in 49 and 67 regions, respectively, of the 83 brain regions examined. The regions affected were those with reported NMDARs, including the neocortex, hippocampus, caudate-putamen, thalamus, substantia nigra, and nucleus accumbens. The increases could be blocked by pretreatment with the specific noncompetitive NMDA antagonist MK-801 ((5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate) (0.3 mg/kg i.p.), as well by a 6-week LiCl diet sufficient to produce plasma and brain lithium concentrations known to be effective in BD. MK-801 alone reduced baseline values for k* in many brain regions. The results show that it is possible to image NMDA signaling via PLA2 activation and AA release in vivo, and that chronic lithium blocks this signaling, consistent with its suggested mechanism of action in BD.


Neurochemical Research | 1999

Dynamics of Docosahexaenoic Acid Metabolism in the Central Nervous System: Lack of Effect of Chronic Lithium Treatment

Michael C. J. Chang; Jane M. Bell; A. D. Purdon; E. G. Chikhale; Eric Grange

Using a method and model developed in our laboratory to quantitatively study brain phospholipid metabolism, in vivo rates of incorporation and turnover of docosahexaenoic acid in brain phospholipids were measured in awake rats. The results suggest that docosahexaenoate incorporation and turnover in brain phospholipids are more rapid than previously assumed and that this rapid turnover dilutes tracer specific activity in brain docoshexaenoyl-CoA pool due to release and recycling of unlabeled fatty acid from phospholipid metabolism. Fractional turnover rates for docosahexaenoate within phosphatidylinositol, choline glycerophospholipids, ethanolamine glycerophospholipids and phosphatidylserine were 17.7, 3.1, 1.2, and 0.2 %.h−1, respectively. Chronic lithium treatment, at a brain level considered to be therapeutic in humans (0.6 μmol.g−1), had no effect on turnover of docosahexaenoic acid in individual brain phospholipids. Consistent with previous studies from our laboratory that chronic lithium decreased the turnover of arachidonic acid within brain phospholipids by up to 80% and attenuated brain phospholipase A2 activity, the lack of effect of lithium on docosahexaenoate recycling and turnover suggests that a target for lithiums action is an arachidonic acid-selective phospholipase A2.


Biological Psychiatry | 2004

Chronic carbamazepine selectively downregulates cytosolic phospholipase A2 expression and cyclooxygenase activity in rat brain

Sandra Ghelardoni; York Tomita; Jane M. Bell; Stanley I. Rapoport; Francesca Bosetti

BACKGROUND Carbamazepine is a mood stabilizer used as monotherapy or as an adjunct to lithium in the treatment of acute mania or the prophylaxis of bipolar disorder. Based on evidence that lithium and valproate, other mood stabilizers, reduce brain arachidonic acid turnover and its conversion via cyclooxygenase to prostaglandin E(2) in rat brain, one possibility is that carbamazepine also targets the arachidonic acid cascade. METHODS To test this hypothesis, carbamazepine was administered to rats by intraperitoneal injection at a daily dose of 25 mg/kg for 30 days. RESULTS Carbamazepine decreased brain phospholipase A(2) activity and cytosolic phospholipase A(2) protein and messenger RNA levels without changing significantly protein and activity levels of calcium-independent phospholipase A(2) or secretory phospholipase A(2). Cyclooxygenase activity was decreased in carbamazepine-treated rats without any change in cyclooxygenase-1 or cyclooxygenase-2 protein levels. Brain prostaglandin E(2) concentration also was reduced. The protein levels of other arachidonic acid metabolizing enzymes, 5-lipoxygenase and cytochrome P450 epoxygenase, were not significantly changed nor was the brain concentration of the 5-lipoxygenase product leukotriene B(4). CONCLUSIONS Carbamazepine downregulates cytosolic phospholipase A(2)-mediated release of arachidonic acid and its subsequent conversion to prostaglandin E(2) by cyclooxygenase. These effects may contribute to its therapeutic actions in bipolar disorder.


Brain Research Bulletin | 2002

Analysis of gene expression with cDNA microarrays in rat brain after 7 and 42 days of oral lithium administration.

Francesca Bosetti; Ruth Seemann; Jane M. Bell; Robert Zahorchak; Elliott Friedman; Stanley I. Rapoport; Pachiappan Manickam

The gene expression profile in rat brain was examined using microarrays in rats fed lithium chloride for 7 days (subacute) or 42 days (chronic). Brain lithium concentrations were 0.39 mM and 0.79 mM (therapeutically relevant), at 7 and 42 days, respectively. Of the 4132 genes represented in the microarrays, 25 genes were downregulated by at least twofold and none was upregulated after 7 days of treatment. Expression of 50 genes was downregulated by at least two-fold at 42 days, without any being upregulated. Lithium treatment for 7 days did not affect at a measurable extent expression of 37 of the 50 genes that were downregulated at 42 days. Genes whose expression was changed at 42 days coded for a number of receptors, protein kinases, transcription and translation factors, markers of energy metabolism, and signal transduction. Thus, chronic lithium at a therapeutically relevant concentration reduced expression of a large number of genes involved in multiple signaling and other pathways, without increasing expression at a comparable extent.

Collaboration


Dive into the Jane M. Bell's collaboration.

Top Co-Authors

Avatar

Stanley I. Rapoport

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lisa Chang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kaizong Ma

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mireille Basselin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael C. J. Chang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Miki Igarashi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

James C. DeMar

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Francesca Bosetti

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mei Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Eric Grange

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge