Network


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

Hotspot


Dive into the research topics where David M. Jacobowitz is active.

Publication


Featured researches published by David M. Jacobowitz.


European Journal of Nuclear Medicine and Molecular Imaging | 2005

PET imaging of brain with the β-amyloid probe, [11C]6-OH-BTA-1, in a transgenic mouse model of Alzheimer’s disease

Hiroshi Toyama; Daniel Ye; Masanori Ichise; Jeih-San Liow; Lisheng Cai; David M. Jacobowitz; John L. Musachio; Jinsoo Hong; Mathew Crescenzo; Dnyanesh Tipre; Jian-Qiang Lu; Sami S. Zoghbi; Douglass Vines; Jurgen Seidel; Kazuhiro Katada; Michael V. Green; Victor W. Pike; Robert M. Cohen; Robert B. Innis

PurposeThe purpose of this study was to evaluate the capacity of [11C]6-OH-BTA-1 and positron emission tomography (PET) to quantify β-amyloid (Aβ) plaques in the Tg2576 mouse model of Alzheimer’s disease (AD).MethodsPET imaging was performed with the NIH ATLAS small animal scanner in six elderly transgenic mice (Tg2576; age 22.0±1.8 months; 23.6±2.6xa0g) overexpressing a mutated form of human β-amyloid precursor protein (APP) known to result in the production of Aβ plaques, and in six elderly wild-type litter mates (age 21.8±1.6 months; 29.5±4.7xa0g). Dynamic PET scans were performed for 30xa0min in each mouse under 1% isoflurane inhalation anesthesia after a bolus injection of 13–46xa0MBq of [11C]6-OH-BTA-1. PET data were reconstructed with 3D OSEM. On the coronal PET image, irregular regions of interest (ROIs) were placed on frontal cortex (FR), parietal cortex (PA), striatum (ST), thalamus (TH), pons (PO), and cerebellum (CE), guided by a mouse stereotaxic atlas. Time–activity curves (TACs) (expressed as percent injected dose per gram normalized to body weight: % ID-kg/g) were obtained for FR, PA, ST, TH, PO, and CE. ROI-to-CE radioactivity ratios were also calculated. Following PET scans, sections of mouse brain prepared from anesthetized and fixative-perfused mice were stained with thioflavin-S.ResultsTACs for [11C]6-OH-BTA-1 in all ROIs peaked early (at 30–55xa0s), with radioactivity washing out quickly thereafter in both transgenic and wild-type mice. Peak uptake in all regions was significantly lower in transgenic mice than in wild-type mice. During the later part of the washout phase (12–30xa0min), the mean FR/CE and PA/CE ratios were higher in transgenic than in wild-type mice (1.06±0.04 vs 0.98±0.07, p=0.04; 1.06±0.09 vs 0.93±0.08 p=0.02) while ST/CE, TH/CE, and PO/CE ratios were not. Ex vivo staining revealed widespread Aβ plaques in cortex, but not in cerebellum of transgenic mice or in any brain regions of wild-type mice.ConclusionMarked reductions in brain uptake of this radioligand in transgenic mice may be due to reduced cerebral blood flow relative to that in wild-type mice. Specific [11C]6-OH-BTA-1 binding to Aβ plaques, if any, is probably very low, as reflected in the small FR/CE and PA/CE ratio differences. FR/CE and PA/CE ratios are considerably higher in AD patients while Aβ plaque densities in 22-month-old transgenic mice may be expected to show essentially the same density as is observed in the AD brain. This implies that the absence of tracer retention in 22-month-old transgenic mice may be due to the smaller number of Aβ plaque binding sites and/or to lower affinity of the binding sites for [11C]6-OH-BTA-1 as compared with AD patients. [11C]6-OH-BTA-1 shows excellent brain uptake in mice.


Life Sciences | 1981

Capsaicin and potassium evoked substance P release from the nucleus tractus solitarius and spinal trigeminal nucleus in vitro

Cinda J. Helke; David M. Jacobowitz; Nguyen B. Thoa

Abstract The nucleus tractus solitarius and the spinal trigeminal nucleus receive peripheral sensory input from substance P containing afferent nerves. This study demonstrates that in vitro depolarization of these nuclei in tissue slices evokes a calcium-dependent efflux of substance P immunoreactivity. Capsaicin (33μM) also elicits substance P release from the nucleus tractus solitarius and spinal trigeminal nucleus but not from the hypothalamus. The occurrence of potassium-stimulated SP release from the two medullary nuclei fulfills one of the criteria for neurotransmitter status. The capsaicin data support the contention that this agent elicits release of substance P from nuclear regions receiving peripheral afferent information in substance P nerves independent of the particular sensory modality served but is ineffective in nonsensory areas.


The Journal of Comparative Neurology | 2006

Abnormal expression of the G-protein-activated inwardly rectifying potassium channel 2 (GIRK2) in hippocampus, frontal cortex, and substantia nigra of Ts65Dn mouse: a model of Down syndrome.

Chie Harashima; David M. Jacobowitz; Jassir Witta; Rosemary C. Borke; Tyler K. Best; Zygmunt Galdzicki

Ts65Dn, a mouse model of Down syndrome (DS), demonstrates abnormal hippocampal synaptic plasticity and behavioral abnormalities related to spatial learning and memory. The molecular mechanisms leading to these impairments have not been identified. In this study, we focused on the G‐protein‐activated inwardly rectifying potassium channel 2 (GIRK2) gene that is highly expressed in the hippocampus region. We studied the expression pattern of GIRK subunits in Ts65Dn and found that GIRK2 was overexpressed in all analyzed Ts65Dn brain regions. Interestingly, elevated levels of GIRK2 protein in the Ts65Dn hippocampus and frontal cortex correlated with elevated levels of GIRK1 protein. This suggests that heteromeric GIRK1‐GIRK2 channels are overexpressed in Ts65Dn hippocampus and frontal cortex, which could impair excitatory input and modulate spike frequency and synaptic kinetics in the affected regions. All GIRK2 splicing isoforms examined were expressed at higher levels in the Ts65Dn in comparison to the diploid hippocampus. The pattern of GIRK2 expression in the Ts65Dn mouse brain revealed by in situ hybridization and immunohistochemistry was similar to that previously reported in the rodent brain. However, in the Ts65Dn mouse a strong immunofluorescent staining of GIRK2 was detected in the lacunosum molecular layer of the CA3 area of the hippocampus. In addition, tyrosine hydroxylase containing dopaminergic neurons that coexpress GIRK2 were more numerous in the substantia nigra compacta and ventral tegmental area in the Ts65Dn compared to diploid controls. In summary, the regional localization and the increased brain levels coupled with known function of the GIRK channel may suggest an important contribution of GIRK2 containing channels to Ts65Dn and thus to DS neurophysiological phenotypes. J. Comp. Neurol. 494:815–833, 2006. Published 2005 Wiley‐Liss, Inc.


Brain Research | 1981

Effect of capsaicin administration to neonatal rats on the substance P content of discrete CNS regions

Cinda J. Helke; Joseph A. DiMicco; David M. Jacobowitz; Irwin J. Kopin

Substance P (SP) levels were determined by radioimmunoassay in microdissected CNS regions of adult animals treated with capsaicin as neonates and of vehicle controls. Capsaicin treatment reduced the SP content of the spinal trigeminal nucleus and the dorsal horn of the spinal cord whereas it had no effect on the SP levels in the ventral horn of the spinal cord, the nucleus tractus solitarius or in midbrain and forebrain areas analyzed.


Molecular Brain Research | 2002

Aspartoacylase is restricted primarily to myelin synthesizing cells in the CNS: therapeutic implications for Canavan disease.

Batool F. Kirmani; David M. Jacobowitz; Abraham T Kallarakal; M.A.A. Namboodiri

Canavan disease is a devastating neurodegenerative childhood disease caused by mutations in aspartoacylase, an enzyme that deacetylates N-acetylaspartate to generate free acetate in the brain. Localization of aspartoacylase in different cell types in the rat brain was examined in an attempt to understand the pathogenesis of Canavan disease. In situ hybridization histochemistry with a riboprobe based on murine aspartoacylase cDNA was used in this study. The hybridization signal was detectable primarily in the myelin-synthesizing cells, namely oligodendroglia. These findings provide strong additional support for insufficient myelin synthesis as the pathogenic basis of Canavan disease and make a compelling case for acetate supplementation as a simple and noninvasive therapy for this fatal disease with no treatment.


Proteomics Clinical Applications | 2007

Protein microarray platforms for clinical proteomics

Harvey B. Pollard; Meera Srivastava; Ofer Eidelman; Catherine Jozwik; Stephen W. Rothwell; Gregory P. Mueller; David M. Jacobowitz; Thomas N. Darling; William B. Guggino; Jerry Wright; Pamela L. Zeitlin; Cloud P. Paweletz

Proteomics for clinical applications is presently in a state of transition. It has become clear that the classical approaches based on 2‐DE and/or MS need to be complemented by different kinds of technologies. The well‐known problems include sample complexity, sensitivity, quantitation, reproducibility, and analysis time. We suggest that the new technologies for clinical proteomics can be supported by antibody‐centric protein microarray platforms. These platforms presently include antibody microarrays and lysate, or reverse capture/reverse phase protein microarrays. Other forms of these arrays are in less mature developmental stages, including ORF and self assembling protein microarrays. Bioinformatic support for interpreting these arrays is becoming more available as the whole field of systems biology begins to mature. The present set of applications for these platforms is profoundly focused on certain common cancers, immunology, and cystic fibrosis. However, we predict that many more disease entities will become studied as knowledge of the power and availability of these platforms becomes more widely established. We anticipate that these platforms will eventually evolve to accommodate label‐free detection technologies, human genome‐scale numbers of analytes, and increases in analytic and bioinformatic speeds.


The Journal of Comparative Neurology | 2010

Nuclear-cytoplasmic localization of acetyl coenzyme A synthetase-1 in the rat brain

Prasanth S. Ariyannur; John R. Moffett; Chikkathur N. Madhavarao; Peethambaran Arun; Nisha Vishnu; David M. Jacobowitz; William C. Hallows; John M. Denu; Aryan M.A. Namboodiri

Acetyl coenzyme A synthetase‐1 (AceCS1) catalyzes the synthesis of acetyl coenzyme A from acetate and coenzyme A and is thought to play diverse roles ranging from fatty acid synthesis to gene regulation. By using an affinity‐purified antibody generated against an 18‐mer peptide sequence of AceCS1 and a polyclonal antibody directed against recombinant AceCS1 protein, we examined the expression of AceCS1 in the rat brain. AceCS1 immunoreactivity in the adult rat brain was present predominantly in cell nuclei, with only light to moderate cytoplasmic staining in some neurons, axons, and oligodendrocytes. Some nonneuronal cell nuclei were very strongly immunoreactive, including those of some oligodendrocytes, whereas neuronal nuclei ranged from unstained to moderately stained. Both antibodies stained some neuronal cell bodies and axons, especially in the hindbrain. AceCS1 immunoreactivity was stronger and more widespread in the brains of 18‐day‐old rats than in adults, with increased expression in oligodendrocytes and neurons, including cortical pyramidal cells. Expression of AceCS1 was substantially up‐regulated in neurons throughout the brain after controlled cortical impact injury. The strong AceCS1 expression observed in the nuclei of CNS cells during brain development and after injury is consistent with a role in nuclear histone acetylation and therefore the regulation of chromatin structure and gene expression. The cytoplasmic staining observed in some oligodendrocytes, especially during postnatal brain development, suggests an additional role in CNS lipid synthesis and myelination. Neuronal and axonal localization implicates AceCS1 in cytoplasmic acetylation reactions in some neurons. J. Comp. Neurol. 518:2952–2977, 2010.


Glia | 2011

Extensive Aspartoacylase Expression in the Rat Central Nervous System

John R. Moffett; Peethambaran Arun; Prasanth S. Ariyannur; James Y. Garbern; David M. Jacobowitz; Aryan M.A. Namboodiri

Aspartoacylase (ASPA) catalyzes deacetylation of N‐acetylaspartate (NAA) to generate acetate and aspartate. Mutations in the gene for ASPA lead to reduced acetate availability in the CNS during development resulting in the fatal leukodystrophy Canavan disease. Highly specific polyclonal antibodies to ASPA were used to examine CNS expression in adult rats. In white matter, ASPA expression was associated with oligodendrocyte cell bodies, nuclei, and some processes, but showed a dissimilar distribution pattern to myelin basic protein and oligodendrocyte specific protein. Microglia expressed ASPA in all CNS regions examined, as did epiplexus cells of the choroid plexus. Pial and ependymal cells and some endothelial cells were ASPA positive, as were unidentified cellular nuclei throughout the CNS. Astrocytes did not express ASPA in their cytoplasm. In some fiber pathways and nerves, particularly in the brainstem and spinal cord, the axoplasm of many neuronal fibers expressed ASPA, as did some neurons. Acetyl coenzyme A synthase immunoreactivity was also observed in the axoplasm of many of the same fiber pathways and nerves. All ASPA‐immunoreactive elements were unstained in brain sections from tremor rats, an ASPA‐null mutant. The strong expression of ASPA in oligodendrocyte cell bodies is consistent with a lipogenic role in myelination. Strong ASPA expression in cell nuclei is consistent with a role for NAA‐derived acetate in nuclear acetylation reactions, including histone acetylation. Expression of ASPA in microglia may indicate a role in lipid synthesis in these cells, whereas expression in axons suggests that some neurons can both synthesize and catabolize NAA.


Brain Research | 2012

Microglia activation along the corticospinal tract following traumatic brain injury in the rat: a neuroanatomical study.

David M. Jacobowitz; Jeffrey Thomas Cole; Dennis P. McDaniel; Harvey B. Pollard; William D. Watson

Traumatic injury to the brain often manifests itself symptomatically and structurally long after the traumatic event. The cellular basis of this complex response is not completely understood. However, we hypothesized that microglia might contribute to the brain-wide process. To test this hypothesis, we employed optical and electron microscopy to study the microglia in rat brains up to 2 months after digitally controlled cortical impact (CCI) to produce traumatic brain injury (TBI). We also used antibodies against ED-1 and Iba-1, respectively, as markers for activated and resting microglia. ED-1 positive microglial cells are observed accompanying the entire corticospinal tract (CST) on the injured side, but not the control, contralateral side of the brain at 2 months. In this case, ED-1 and Iba-1 were observed to co-localize uniquely on the injured side of the brain. At earlier times following CCI, ultrastructural studies reveal that microglial cells have very irregular shapes and have many processes that intermingle with degenerating nerve axons of the CST in the hindbrain pyramids. These cells appear to be engulfing degenerating myelinated axons. The debris within the cells is converted to lipofuscin, the antigen for the ED-1 antibody, and remains in the cell cytoplasm throughout the life of the cell. We conclude, as hypothesized, that microglia are critical cellular components. Based on observed close association with myelin degeneration, interdigitating activated microglia may be contributing to damage control. Finally, based on the close neuroanatomical relationship between the lesioned corticospinal tract and the wide distribution of activated microglia, primary signals from CST neurons per se, may be directing microglial responses along the entire damaged rat neuroaxis. The role of persistent activation of microglia has not been determined.


Free Radical Biology and Medicine | 2015

Protein carbonylation after traumatic brain injury: cell specificity, regional susceptibility, and gender differences

Rachel C. Lazarus; John E. Buonora; David M. Jacobowitz; Gregory P. Mueller

Protein carbonylation is a well-documented and quantifiable consequence of oxidative stress in several neuropathologies, including multiple sclerosis, Alzheimer׳s disease, and Parkinson׳s disease. Although oxidative stress is a hallmark of traumatic brain injury (TBI), little work has explored the specific neural regions and cell types in which protein carbonylation occurs. Furthermore, the effect of gender on protein carbonylation after TBI has not been studied. The present investigation was designed to determine the regional and cell specificity of TBI-induced protein carbonylation and how this response to injury is affected by gender. Immunohistochemistry was used to visualize protein carbonylation in the brains of adult male and female Sprague-Dawley rats subjected to controlled cortical impact (CCI) as an injury model of TBI. Cell-specific markers were used to colocalize the presence of carbonylated proteins in specific cell types, including astrocytes, neurons, microglia, and oligodendrocytes. Results also indicated that the injury lesion site, ventral portion of the dorsal third ventricle, and ventricular lining above the median eminence showed dramatic increases in protein carbonylation after injury. Specifically, astrocytes and limited regions of ependymal cells adjacent to the dorsal third ventricle and the median eminence were most susceptible to postinjury protein carbonylation. However, these patterns of differential susceptibility to protein carbonylation were gender dependent, with males showing significantly greater protein carbonylation at sites distant from the lesion. Proteomic analyses were also conducted and determined that the proteins most affected by carbonylation in response to TBI include glial fibrillary acidic protein, dihydropyrimidase-related protein 2, fructose-bisphosphate aldolase C, and fructose-bisphosphate aldolase A. Many other proteins, however, were not carbonylated by CCI. These findings indicate that there is both regional and protein specificity in protein carbonylation after TBI. The marked increase in carbonylation seen in ependymal layers distant from the lesion suggests a mechanism involving the transmission of a cerebral spinal fluid-borne factor to these sites. Furthermore, this process is affected by gender, suggesting that hormonal mechanisms may serve a protective role against oxidative stress.

Collaboration


Dive into the David M. Jacobowitz's collaboration.

Top Co-Authors

Avatar

Harvey B. Pollard

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Clifton L. Dalgard

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Gregory P. Mueller

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Robert J. Ursano

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Catherine Jozwik

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

John E. Buonora

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Meera Srivastava

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Ofer Eidelman

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Rachel C. Lazarus

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Ann M. Marini

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge