Network


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

Hotspot


Dive into the research topics where G. Jean Harry is active.

Publication


Featured researches published by G. Jean Harry.


Pharmacology & Therapeutics | 2013

Microglia during development and aging.

G. Jean Harry

Microglia are critical nervous system-specific cells influencing brain development, maintenance of the neural environment, response to injury, and repair. They contribute to neuronal proliferation and differentiation, pruning of dying neurons, synaptic remodeling and clearance of debris and aberrant proteins. Colonization of the brain occurs during gestation with an expansion following birth with localization stimulated by programmed neuronal death, synaptic pruning, and axonal degeneration. Changes in microglia phenotype relate to cellular processes including specific neurotransmitter, pattern recognition, or immune-related receptor activation. Upon activation, microglia cells have the capacity to release a number of substances, e.g., cytokines, chemokines, nitric oxide, and reactive oxygen species, which could be detrimental or beneficial to the surrounding cells. With aging, microglia shift their morphology and may display diminished capacity for normal functions related to migration, clearance, and the ability to shift from a pro-inflammatory to an anti-inflammatory state to regulate injury and repair. This shift in microglia potentially contributes to increased susceptibility and neurodegeneration as a function of age. In the current review, information is provided on the colonization of the brain by microglia, the expression of various pattern recognition receptors to regulate migration and phagocytosis, and the shift in related functions that occur in normal aging.


International Journal of Environmental Research and Public Health | 2011

Features of Microglia and Neuroinflammation Relevant to Environmental Exposure and Neurotoxicity

Andrew D. Kraft; G. Jean Harry

Microglia are resident cells of the brain involved in regulatory processes critical for development, maintenance of the neural environment, injury and repair. They belong to the monocytic-macrophage lineage and serve as brain immune cells to orchestrate innate immune responses; however, they are distinct from other tissue macrophages due to their relatively quiescent phenotype and tight regulation by the CNS microenvironment. Microglia actively survey the surrounding parenchyma and respond rapidly to changes such that any disruption to neural architecture or function can contribute to the loss in regulation of the microglia phenotype. In many models of neurodegeneration and neurotoxicity, early events of synaptic degeneration and neuronal loss are accompanied by an inflammatory response including activation of microglia, perivascular monocytes, and recruitment of leukocytes. In culture, microglia have been shown to be capable of releasing several potentially cytotoxic substances, such as reactive oxygen intermediates, nitric oxide, proteases, arachidonic acid derivatives, excitatory amino acids, and cytokines; however, they also produce various neurotrophic factors and quench damage from free radicals and excitotoxins. As the primary source for pro-inflammatory cytokines, microglia are implicated as pivotal mediators of neuroinflammation and can induce or modulate a broad spectrum of cellular responses. Neuroinflammation should be considered as a balanced network of processes whereby subtle modifications can shift the cells toward disparate outcomes. For any evaluation of neuroinflammation and microglial responses, within the framework of neurotoxicity or degeneration, one key question in determining the consequence of neuroinflammation is whether the response is an initiating event or the consequence of tissue damage. As examples of environmental exposure-related neuroinflammation in the literature, we provide an evaluation of data on manganese and diesel exhaust particles.


Neurotoxicology | 2012

Microglia in the developing brain: a potential target with lifetime effects

G. Jean Harry; Andrew D. Kraft

Microglia are a heterogenous group of monocyte-derived cells serving multiple roles within the brain, many of which are associated with immune and macrophage like properties. These cells are known to serve a critical role during brain injury and to maintain homeostasis; yet, their defined roles during development have yet to be elucidated. Microglial actions appear to influence events associated with neuronal proliferation and differentiation during development, as well as, contribute to processes associated with the removal of dying neurons or cellular debris and management of synaptic connections. These long-lived cells display changes during injury and with aging that are critical to the maintenance of the neuronal environment over the lifespan of the organism. These processes may be altered by changes in the colonization of the brain or by inflammatory events during development. This review addresses the role of microglia during brain development, both structurally and functionally, as well as the inherent vulnerability of the developing nervous system. A framework is presented considering microglia as a critical nervous system-specific cell that can influence multiple aspects of brain development (e.g., vascularization, synaptogenesis, and myelination) and have a long term impact on the functional vulnerability of the nervous system to a subsequent insult, whether environmental, physical, age-related, or disease-related.


Journal of Neurochemistry | 2002

Cellular localization and temporal elevation of tumor necrosis factor-α, interleukin-1α, and transforming growth factor-β1 mRNA in hippocampal injury response induced by trimethyltin

Alessandra Bruccoleri; Hayes W. Brown; G. Jean Harry

Abstract: In certain pathologic states, cytokine production may become spatially and temporally dysregulated, leading to their inappropriate production and potentially detrimental consequences. Tumor necrosis factor‐α (TNF‐α), interleukin (IL)‐1, IL‐6, and transforming growth factor‐β (TGF‐β) mediate a range of host responses affecting multiple cell types. To study the role of cytokines in the early stages of brain injury, we examined alterations in the 17‐day‐old mouse hippocampus during trimethyltin‐induced neurodegeneration characterized by neuronal necrosis, microglia activation in the dentate, and astrocyte reactivity throughout the hippocampus. By 24 h after dosing, elevations in mRNA levels for TNF‐α, IL‐1α, IL‐1β, and IL‐6 mRNA were seen. TGF‐β1 mRNA was elevated at 72 h. In situ hybridization showed that TNF‐α and IL‐1α were localized to the microglia, whereas TGF‐β1 was expressed predominantly in hippocampal pyramidal cells. Intercellular adhesion molecule‐1, EB‐22, Mac‐1, and glial fibrillary acidic protein mRNA levels were elevated within the first 3 days of exposure in the absence of increased inducible nitric oxide synthetase and interferon‐γ mRNA. These data suggest that pro‐inflammatory cytokines contribute to the progression and pattern of neuronal degeneration in the hippocampus.


Journal of Neurochemistry | 2004

Rat brain arachidonic acid metabolism is increased by a 6-day intracerebral ventricular infusion of bacterial lipopolysaccharide: Neuroinflammation alters brain arachidonic acid metabolism

Thad A. Rosenberger; Nelly E. Villacreses; Jonathan T. Hovda; Francesca Bosetti; Gayani R. Weerasinghe; Robert N. Wine; G. Jean Harry; Stanley I. Rapoport

In a rat model of acute neuroinflammation, produced by a 6‐day intracerebral ventricular infusion of bacterial lipopolysaccharide (LPS), we measured brain activities and protein levels of three phospholipases A2 (PLA2) and of cyclo‐oxygenase‐1 and ‐2, and quantified other aspects of brain phospholipid and fatty acid metabolism. The 6‐day intracerebral ventricular infusion increased lectin‐reactive microglia in the cerebral ventricles, pia mater, and the glial membrane of the cortex and resulted in morphological changes of glial fibrillary acidic protein (GFAP)‐positive astrocytes in the cortical mantel and areas surrounding the cerebral ventricles. LPS infusion increased brain cytosolic and secretory PLA2 activities by 71% and 47%, respectively, as well as the brain concentrations of non‐esterified linoleic and arachidonic acids, and of prostaglandins E2 and D2. LPS infusion also increased rates of incorporation and turnover of arachidonic acid in phosphatidylethanolamine, plasmenylethanolamine, phosphatidylcholine, and plasmenylcholine by 1.5‐ to 2.8‐fold, without changing these rates in phosphatidylserine or phosphatidylinositol. These observations suggest that selective alterations in brain arachidonic acid metabolism involving cytosolic and secretory PLA2 contribute to early pathology in neuroinflammation.


Neurotoxicology | 2003

Dentate gyrus: alterations that occur with hippocampal injury.

G. Jean Harry; Christian Lefebvre d’Hellencourt

Injury to the brain usually manifests not in a diffuse uniform manner but rather with selective sites of damage indicative of differential vulnerability. This question of neuronal susceptibility has been one of major interest both in disease processes as well as damage induced by environmental factors. For experimental examination, brain structures with obvious neuronal subpopulations and organization such as the cerebellum and the hippocampus have offered the most promise. In the hippocampus distinct neuronal populations exist that demonstrate differential vulnerability to various forms of insult including ischemia, excitotoxicity, and environmental factors. The more recent data regarding the presence of neuronal progenitor cells in the subgranular zone of the dentate offers the opportunity to expand such experimental examination to the process of injury-induced neurogenesis. Thus, more recent studies have expanded the examination of the hippocampus to include models of damage to the dentate neurons in addition to the highly vulnerable pyramidal neurons. A number of these models are presented for both human disease and experimental animal conditions. Examination of the responses between these distinct cell populations offers the potential for understanding factors that are critical in neuronal death and survival.


Brain Behavior and Immunity | 2011

Voluntary exercise protects hippocampal neurons from trimethyltin injury: Possible role of interleukin-6 to modulate tumor necrosis factor receptor-mediated neurotoxicity

Jason A. Funk; Julia M. Gohlke; Andrew D. Kraft; Christopher A. McPherson; Jennifer B. Collins; G. Jean Harry

In the periphery, exercise induces interleukin (IL)-6 to downregulate tumor necrosis factor (TNF), elevate interleukin-1 receptor antagonist (IL-1RA), decreasing inflammation. Exercise also offers neuroprotection and facilitates brain repair. IL-6 production in the hippocampus following exercise suggests the potential of a similar protective role as in the periphery to down-regulate TNFα and inflammation. Using a chemical-induced model of hippocampal dentate granule cell death (trimethyltin, TMT 2.4 mg/kg, ip) dependent upon TNF receptor signaling, we demonstrate neuroprotection in mice with 2 weeks access to running wheel. Exercise attenuated neuronal death and diminished elevations in TNFα, TNF receptor 1, myeloid differentiation primary response gene (MyD) 88, transforming growth factor β, chemokine (C-C motif) ligand 2 (CCL2), and CCL3. Elevated mRNA levels for IL-1α, IL-1RA, occurred with injury and protection. mRNA and protein levels of IL-6 and neuronal expression of IL-6 receptor α, were elevated with injury and protection. Microarray pathway analysis supported an up-regulation of TNFα cell death signaling pathways with TMT and inhibition by exercise. IL-6 pathway recruitment occurred in both conditions. IL-6 downstream signal events differed in the level of STAT3 activation. Exercise did not increase mRNA levels of brain derived neurotrophic factor, nerve growth factor, or glial derived neurotrophic factor. In IL-6 deficient mice, exercise did not attenuate TMT-induced tremor and a diminished level of neuroprotection was observed. These data suggest a contributory role for IL-6 induced by exercise for neuroprotection in the CNS similar to that seen in the periphery.


Neurotoxicology | 2009

Heterogeneity of microglia and TNF signaling as determinants for neuronal death or survival

Andrew D. Kraft; Christopher A. McPherson; G. Jean Harry

Microglia do not constitute a single, uniform cell population, but rather comprise cells with varied phenotypes, some which are beneficial and others that may require active regulatory control. Thus, gaining a better understanding of the heterogeneity of resident microglia responses will contribute to any interpretation regarding the impact of any such response in the brain. Microglia are the primary source of the pro-inflammatory cytokine, tumor necrosis factor (TNF) that can initiate various effects through the activation of membrane receptors. The TNF p55 receptor contains a death domain and activation normally leads to cellular apoptosis; however, under specific conditions, receptor activation can also lead to the activation of NF-kappaB and contribute to cell survival. These divergent outcomes have been linked to receptor localization with receptor internalization leading to cell death and membrane localization supporting cell survival. A second TNF receptor, TNF p75 receptor, is normally linked to cell growth and survival, however, it can cooperate with the p55 receptor and contribute to cell death. Thus, while an elevation in TNFalpha in the brain is often considered an indicator of microglia activation and neuroinflammation, a number of factors come into play to determine the final outcome. Data are reviewed demonstrating that heterogeneity in morphological response of microglia and the expression of TNFalpha and TNF receptors are critical in identifying and characterizing neurotoxic events as they relate to neuroinflammation, neuronal damage and in stimulating neuroprotection.


Journal of Neurochemistry | 2008

Tumor necrosis factor p55 and p75 receptors are involved in chemical-induced apoptosis of dentate granule neurons

G. Jean Harry; Christian Lefebvre d’Hellencourt; Christopher A. McPherson; Jason A. Funk; Mineyoshi Aoyama; Robert N. Wine

Localized tumor necrosis factor‐α (TNFα) elevation has diverse effects in brain injury often attributed to signaling via TNFp55 or TNFp75 receptors. Both dentate granule cells and CA pyramidal cells express TNF receptors (TNFR) at low levels in a punctate pattern. Using a model to induce selective death of dentate granule cells (trimethyltin; 2 mg/kg, i.p.), neuronal apoptosis [terminal deoxynucleotidyl transferase‐mediated dUTP‐biotin in situ end labeling, active caspase 3 (AC3)] was accompanied by amoeboid microglia and elevated TNFα mRNA levels. TNFp55R (55 kDa type‐1 TNFR) and TNFp75R (75 kDa type‐2 TNFR) immunoreactivity in AC3+ neurons displayed a pattern suggestive of receptor internalization and a temporal sequence of expression of TNFp55R followed by TNFp75R associated with the progression of apoptosis. A distinct ramified microglia response occurred around CA1 neurons and healthy dentate neurons that displayed an increase in the normal punctate pattern of TNFRs. Neuronal damage was decreased with i.c.v. injection of TNFα antibody and in TNFp55R−/−p75R−/− mice that showed higher constitutive mRNA levels for interleukin (IL‐1α), macrophage inflammatory protein 1‐α (MIP‐1α), TNFα, transforming growth factor β1, Fas, and TNFRSF6‐assoicated via death domain (FADD). TNFp75R−/− mice showed exacerbated injury and elevated mRNA levels for IL‐1α, MIP‐1α, and TNFα. In TNFp55R−/− mice, constitutive mRNA levels for TNFα, IL‐6, caspase 8, FADD, and Fas‐associated phosphatase were higher; IL‐1α, MIP‐1α, and transforming growth factor β1 lower. The mice displayed exacerbated neuronal death, delayed microglia response, increased FADD and TNFp75R mRNA levels, and co‐expression of TNFp75R in AC3+ neurons. The data demonstrate TNFR‐mediated apoptotic death of dentate granule neurons utilizing both TNFRs and suggest a TNFp75R‐mediated apoptosis in the absence of normal TNFp55R activity.


Environmental Health Perspectives | 2007

Ontogenetic alterations in molecular and structural correlates of dendritic growth after developmental exposure to polychlorinated biphenyls

Pamela J. Lein; Dongren Yang; Adam D. Bachstetter; Hugh A. Tilson; G. Jean Harry; Ronald F. Mervis; Prasada Rao S. Kodavanti

Objective Perinatal exposure to polychlorinated biphenyls (PCBs) is associated with decreased IQ scores, impaired learning and memory, psychomotor difficulties, and attentional deficits in children. It is postulated that these neuropsychological deficits reflect altered patterns of neuronal connectivity. To test this hypothesis, we examined the effects of developmental PCB exposure on dendritic growth. Methods Rat dams were gavaged from gestational day 6 through postnatal day (PND) 21 with vehicle (corn oil) or the commercial PCB mixture Aroclor 1254 (6 mg/kg/day). Dendritic growth and molecular markers were examined in pups during development. Results Golgi analyses of CA1 hippocampal pyramidal neurons and cerebellar Purkinge cells indicated that developmental exposure to PCBs caused a pronounced age-related increase in dendritic growth. Thus, even though dendritic lengths were significantly attenuated in PCB-treated animals at PND22, the rate of growth was accelerated at later ages such that by PND60, dendritic growth was comparable to or even exceeded that observed in vehicle controls. Quantitative reverse transcriptase polymerase chain reaction analyses demonstrated that from PND4 through PND21, PCBs generally increased expression of both spinophilin and RC3/neurogranin mRNA in the hippocampus, cerebellum, and cortex with the most significant increases observed in the cortex. Conclusions This study demonstrates that developmental PCB exposure alters the ontogenetic profile of dendritogenesis in critical brain regions, supporting the hypothesis that disruption of neuronal connectivity contributes to neuropsychological deficits seen in exposed children.

Collaboration


Dive into the G. Jean Harry's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew D. Kraft

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alessandra Bruccoleri

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert N. Wine

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susan Brunssen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge