Network


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

Hotspot


Dive into the research topics where Michelle L. Block is active.

Publication


Featured researches published by Michelle L. Block.


Nature Reviews Neuroscience | 2007

Microglia-mediated neurotoxicity: uncovering the molecular mechanisms

Michelle L. Block; Luigi Zecca; Jau Shyong Hong

Mounting evidence indicates that microglial activation contributes to neuronal damage in neurodegenerative diseases. Recent studies show that in response to certain environmental toxins and endogenous proteins, microglia can enter an overactivated state and release reactive oxygen species (ROS) that cause neurotoxicity. Pattern recognition receptors expressed on the microglial surface seem to be one of the primary, common pathways by which diverse toxin signals are transduced into ROS production. Overactivated microglia can be detected using imaging techniques and therefore this knowledge offers an opportunity not only for early diagnosis but, importantly, for the development of targeted anti-inflammatory therapies that might slow or halt the progression of neurodegenerative disease.


Glia | 2007

Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration

Liya Qin; Xuefei Wu; Michelle L. Block; Yuxin Liu; George R. Breese; Jau Shyong Hong; Darin J. Knapp; Fulton T. Crews

Inflammation is implicated in the progressive nature of neurodegenerative diseases, such as Parkinsons disease, but the mechanisms are poorly understood. A single systemic lipopolysaccharide (LPS, 5 mg/kg, i.p.) or tumor necrosis factor alpha (TNFα, 0.25 mg/kg, i.p.) injection was administered in adult wild‐type mice and in mice lacking TNFα receptors (TNF R1/R2−/−) to discern the mechanisms of inflammation transfer from the periphery to the brain and the neurodegenerative consequences. Systemic LPS administration resulted in rapid brain TNFα increase that remained elevated for 10 months, while peripheral TNFα (serum and liver) had subsided by 9 h (serum) and 1 week (liver). Systemic TNFα and LPS administration activated microglia and increased expression of brain pro‐inflammatory factors (i.e., TNFα, MCP‐1, IL‐1β, and NF‐κB p65) in wild‐type mice, but not in TNF R1/R2−/− mice. Further, LPS reduced the number of tyrosine hydroxylase‐immunoreactive neurons in the substantia nigra (SN) by 23% at 7‐months post‐treatment, which progressed to 47% at 10 months. Together, these data demonstrate that through TNFα, peripheral inflammation in adult animals can: (1) activate brain microglia to produce chronically elevated pro‐inflammatory factors; (2) induce delayed and progressive loss of DA neurons in the SN. These findings provide valuable insight into the potential pathogenesis and self‐propelling nature of Parkinsons disease.


The FASEB Journal | 2005

Aggregated α-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease

Wei Zhang; Tongguang Wang; Zhong Pei; David S. Miller; Xuefei Wu; Michelle L. Block; Belinda Wilson; Wanqin Zhang; Yong Zhou; Jau Shyong Hong; Jing Zhang

A growing body of evidence indicates that an inflammatory process in the substantia nigra, characterized by activation of resident microglia, likely either initiates or aggravates nigral neurodegeneration in Parkinsons disease (PD). To study the mechanisms by which nigral microglia are activated in PD, the potential role of α‐synuclein (a major component of Lewy bodies that can cause neurodegeneration when aggregated) in microglial activation was investigated. The results demonstrated that in a primary mesencephalic neuron‐glia culture system, extracellular aggregated human α‐synuclein indeed activated microglia; microglial activation enhanced dopaminergic neurodegeneration induced by aggregated α‐synuclein. Furthermore, microglial enhancement of α‐synuclein‐mediated neurotoxicity depended on phagocytosis of α‐synuclein and activation of NADPH oxidase with production of reactive oxygen species. These results suggest that nigral neuronal damage, regardless of etiology, may release aggregated α‐synuclein into substantia nigra, which activates microglia with production of proinflammatory mediators, thereby leading to persistent and progressive nigral neurodegeneration in PD. Finally, NADPH oxidase could be an ideal target for potential pharmaceutical intervention, given that it plays a critical role in α‐synuclein‐mediated microglial activation and associated neurotoxicity.—Zhang, W., Wang, T., Pei, Z., Miller, D. S., Wu, X., Block, M. L., Wilson, B., Zhang, W., Zhou, Y., Hong, J. S., Zhang, J. Aggregated α‐synuclein activates microglia: a process leading to disease progression in Parkinsons disease. FASEB J. 19, 533–542 (2005)


Trends in Neurosciences | 2009

Air pollution: mechanisms of neuroinflammation and CNS disease

Michelle L. Block; Lilian Calderón-Garcidueñas

Air pollution has been implicated as a chronic source of neuroinflammation and reactive oxygen species (ROS) that produce neuropathology and central nervous system (CNS) disease. Stroke incidence and Alzheimers and Parkinsons disease pathology are linked to air pollution. Recent reports reveal that air pollution components reach the brain; systemic effects that impact lung and cardiovascular disease also impinge upon CNS health. While mechanisms driving air pollution-induced CNS pathology are poorly understood, new evidence suggests that microglial activation and changes in the blood-brain barrier are key components. Here we summarize recent findings detailing the mechanisms through which air pollution reaches the brain and activates the resident innate immune response to become a chronic source of pro-inflammatory factors and ROS, culminating in CNS disease.


Neurotherapeutics | 2010

Microglial Activation and Chronic Neurodegeneration

Melinda E. Lull; Michelle L. Block

SummaryMicroglia, the resident innate immune cells in the brain, have long been implicated in the pathology of neurode-generative diseases. Accumulating evidence points to activated microglia as a chronic source of multiple neurotoxic factors, including tumor necrosis factor-α, nitric oxide, interleukin-1β, and reactive oxygen species (ROS), driving progressive neuron damage. Microglia can become chronically activated by either a single stimulus (e.g., lipopolysaccharide or neuron damage) or multiple stimuli exposures to result in cumulative neuronal loss with time. Although the mechanisms driving these phenomena are just beginning to be understood, reactive microgliosis (the microglial response to neuron damage) and ROS have been implicated as key mechanisms of chronic and neurotoxic microglial activation, particularly in the case of Parkinson’s disease. We review the mechanisms of neurotoxicity associated with chronic microglial activation and discuss the role of neuronal death and microglial ROS driving the chronic and toxic microglial phenotype.


The FASEB Journal | 2004

Nanometer size diesel exhaust particles are selectively toxic to dopaminergic neurons: the role of microglia, phagocytosis, and NADPH oxidase

Michelle L. Block; Xufeng Wu; Zhong Pei; Guanhan Li; Tongguang Wang; Liya Qin; Belinda Wilson; Jun Yang; Jau-Shyong Hong; Bellina Veronesi

The contributing role of environmental factors to the development of Parkinson’s disease has become increasingly evident. We report that mesencephalic neuron‐glia cultures treated with diesel exhaust particles (DEP; 0.22 µM) (5–50 µg/ml) resulted in a dose‐dependent decrease in dopaminergic (DA) neurons, as determined by DA‐uptake assay and tyrosine‐hydroxylase immunocytochemistry (ICC). The selective toxicity of DEP for DA neurons was demonstrated by the lack of DEP effect on both GABA uptake and Neu‐N immunoreactive cell number. The critical role of microglia was demonstrated by the failure of neuron‐enriched cultures to exhibit DEP‐induced DA neurotoxicity, where DEP‐induced DA neuron death was reinstated with the addition of microglia to neuron‐enriched cultures. OX‐42 ICC staining of DEP treated neuron‐glia cultures revealed changes in microglia morphology indicative of activation. Intracellular reactive oxygen species and superoxide were produced from enriched‐microglia cultures in response to DEP. Neuron‐glia cultures from NADPH oxidase deficient (PHOX−/−) mice were insensitive to DEP neurotoxicity when compared with control mice (PHOX+/+). Cytochalasin D inhibited DEP‐induced superoxide production in enriched‐microglia cultures, implying that DEP must be phagocytized by microglia to produce superoxide. Together, these in vitro data indicate that DEP selectively damages DA neurons through the phagocytic activation of microglial NADPH oxidase and consequent oxidative insult.


Biochemical Society Transactions | 2007

Chronic microglial activation and progressive dopaminergic neurotoxicity

Michelle L. Block; Jau Shyong Hong

PD (Parkinsons disease) is characterized by the selective and progressive loss of DA neurons (dopaminergic neurons) in the substantia nigra. Inflammation and activation of microglia, the resident innate immune cell in the brain, have been strongly linked to neurodegenerative diseases, such as PD. Microglia can respond to immunological stimuli and neuronal death to produce a host of toxic factors, including cytokines and ROS (reactive oxygen species). Microglia can also become persistently activated after a single stimulus and maintain the elevated production of both cytokines and ROS, long after the instigating stimulus is gone. Current reports suggest that this chronic microglial activation may be fuelled by either dying/damaged neurons or autocrine and paracrine signals from local glial cells, such as cytokines. Here, we review proposed mechanisms responsible for chronic neuroinflammation and explain the interconnected relationship between deleterious microglial activation, DA neuron damage and neurodegenerative disease.


The International Journal of Neuropsychopharmacology | 2008

Histone deacetylase inhibitors up-regulate astrocyte GDNF and BDNF gene transcription and protect dopaminergic neurons

Xuefei Wu; Po-See Chen; Shannon Dallas; Belinda Wilson; Michelle L. Block; Chao Chuan Wang; Harriet Kinyamu; Nick Z. Lu; Xi Gao; Yan Leng; De Maw Chuang; Wanqin Zhang; Ru-Band Lu; Jau Shyong Hong

Parkinsons disease (PD) is characterized by the selective and progressive loss of dopaminergic (DA) neurons in the midbrain substantia nigra. Currently, available treatment is unable to alter PD progression. Previously, we demonstrated that valproic acid (VPA), a mood stabilizer, anticonvulsant and histone deacetylase (HDAC) inhibitor, increases the expression of glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) in astrocytes to protect DA neurons in midbrain neuron-glia cultures. The present study investigated whether these effects are due to HDAC inhibition and histone acetylation. Here, we show that two additional HDAC inhibitors, sodium butyrate (SB) and trichostatin A (TSA), mimic the survival-promoting and protective effects of VPA on DA neurons in neuron-glia cultures. Similar to VPA, both SB and TSA increased GDNF and BDNF transcripts in astrocytes in a time-dependent manner. Furthermore, marked increases in GDNF promoter activity and promoter-associated histone H3 acetylation were noted in astrocytes treated with all three compounds, where the time-course for acetylation was similar to that for gene transcription. Taken together, our results indicate that HDAC inhibitors up-regulate GDNF and BDNF expression in astrocytes and protect DA neurons, at least in part, through HDAC inhibition. This study indicates that astrocytes may be a critical neuroprotective mechanism of HDAC inhibitors, revealing a novel target for the treatment of psychiatric and neurodegenerative diseases.


Neurotoxicology | 2012

The outdoor air pollution and brain health workshop

Michelle L. Block; Alison Elder; Richard L. Auten; Staci D. Bilbo; Honglei Chen; Jiu Chiuan Chen; Deborah A. Cory-Slechta; Daniel L. Costa; David Diaz-Sanchez; David C. Dorman; Diane R. Gold; Kimberly A. Gray; Hueiwang Anna Jeng; Joel D. Kaufman; Michael T. Kleinman; Annette Kirshner; Cindy P. Lawler; David S. Miller; Srikanth S. Nadadur; Beate Ritz; Erin O. Semmens; Leonardo H. Tonelli; Bellina Veronesi; Robert O. Wright; Rosalind J. Wright

Accumulating evidence suggests that outdoor air pollution may have a significant impact on central nervous system (CNS) health and disease. To address this issue, the National Institute of Environmental Health Sciences/National Institute of Health convened a panel of research scientists that was assigned the task of identifying research gaps and priority goals essential for advancing this growing field and addressing an emerging human health concern. Here, we review recent findings that have established the effects of inhaled air pollutants in the brain, explore the potential mechanisms driving these phenomena, and discuss the recommended research priorities/approaches that were identified by the panel.


The FASEB Journal | 2008

Diesel exhaust particles induce oxidative stress, proinflammatory signaling, and P-glycoprotein up-regulation at the blood-brain barrier

Anika M.S. Hartz; Björn Bauer; Michelle L. Block; Jau Shyong Hong; David S. Miller

Here, we report that diesel exhaust particles (DEPs), a major constituent of urban air pollution, affect blood‐brain barrier function at the tissue, cellular, and molecular levels. Isolated rat brain capillaries exposed to DEPs showed increased expression and transport activity of the key drug efflux transporter, P‐glycoprotein (6 h EC50 was ~5 μg/ml). Upregulation of P‐glycoprotein was abolished by blocking transcription or protein synthesis. Inhibition of NADPH oxidase or pretreatment of capillaries with radical scavengers ameliorated DEP‐induced P‐glycoprotein up‐regulation, indicating a role for reactive oxygen species in signaling. DEP exposure also increased brain capillary tumor necrosis factor‐α (TNF‐α) levels. DEP‐induced P‐glycoprotein up‐regulation was abolished when TNF‐receptor 1 (TNF‐R1) was blocked and was not evident in experiments with capillaries from TNF‐R1 knockout mice. Inhibition of JNK, but not NF‐κB, blocked DEP‐induced P‐glycoprotein up‐regulation, indicating a role for AP‐1 in the signaling pathway. Consistent with this, DEPs increased phosphorylation of c‐jun. Together, our results show for the first time that a component of air pollution, DEPs, alters blood‐brain barrier function through oxidative stress and proinflammatory cytokine production. These experiments disclose a novel blood‐brain barrier signaling pathway, with clear implications for environmental toxicology, CNS pathology, and the pharmacotherapy of CNS disorders.—Hartz, A. M. S., Bauer, B., Block, M. L., Hong, J.‐S., Miller, D.‐S. Diesel exhaust particles induce oxidative stress, proinflammatory signaling, and P‐glycoprotein up‐regulation at the blood‐brain barrier. FASEB J. 22, 2723–2733 (2008)

Collaboration


Dive into the Michelle L. Block's collaboration.

Top Co-Authors

Avatar

Jau Shyong Hong

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Belinda Wilson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shannon Levesque

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Jau-Shyong Hong

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Liya Qin

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Wei Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tongguang Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xuefei Wu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhong Pei

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David S. Miller

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge