Network


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

Hotspot


Dive into the research topics where Junming Wang is active.

Publication


Featured researches published by Junming Wang.


Neuropsychopharmacology | 2011

The reduction of R1, a novel repressor protein for monoamine oxidase A, in major depressive disorder.

Shakevia Johnson; Craig A. Stockmeier; Jeffrey H. Meyer; Mark C. Austin; Paul R. Albert; Junming Wang; Warren L. May; Grazyna Rajkowska; James C. Overholser; George Jurjus; Lesa Dieter; Chandra Johnson; Donald B. Sittman; Xiao-Ming Ou

The novel transcriptional repressor protein, R1 (JPO2/CDCA7L/RAM2), inhibits monoamine oxidase A (MAO A) gene expression and influences cell proliferation and survival. MAO A is implicated in several neuropsychiatric illnesses and highly elevated in major depressive disorder (MDD); however, whether R1 is involved in these disorders is unknown. This study evaluates the role of R1 in depressed subjects either untreated or treated with antidepressant drugs. R1 protein levels were determined in the postmortem prefrontal cortex of 18 untreated MDD subjects and 12 medicated MDD subjects compared with 18 matched psychiatrically normal control subjects. Western blot analysis showed that R1 was significantly decreased by 37.5% (p<0.005) in untreated MDD subjects. The R1 level in medicated MDD subjects was also significantly lower (by 30%; p<0.05) compared with control subjects, but was not significantly different compared with untreated MDD subjects. Interestingly, the reduction in R1 was significantly correlated with an increase (approximately 40%; p<0.05) in MAO A protein levels within the MDD groups compared with controls. Consistent with the change in MAO A protein expression, the MAO A catalytic activity was significantly greater in both MDD groups compared with controls. These results suggest that reduced R1 may lead to elevated MAO A levels in untreated and treated MDD subjects; moreover, the reduction of R1 has been implicated in apoptotic cell death and apoptosis has also been observed in the brains of MDD subjects. Therefore, modulation of R1 levels may provide a new therapeutic target in the development of more effective strategies to treat MDD.


Journal of Biological Chemistry | 2012

Mechanistic Role for a Novel Glucocorticoid-KLF11 (TIEG2) Protein Pathway in Stress-induced Monoamine Oxidase A Expression

Matthew Grunewald; Shakevia Johnson; Deyin Lu; Zhe Wang; Gwen Lomberk; Paul R. Albert; Craig A. Stockmeier; Jeffrey H. Meyer; Raul Urrutia; Klaus A. Miczek; Mark C. Austin; Junming Wang; Ian A. Paul; William L. Woolverton; Seungmae Seo; Donald B. Sittman; Xiao Ming Ou

Background: The function of KLF11/TIEG2 under stressful conditions is undefined. Results: KLF11 increases brain MAO expression through its promoter and a chromatin partner, which can be enhanced by stress. Conclusion: This is the first elucidation of mechanisms underlying stress-induced KLF11-MAO up-regulation. Significance: This novel KLF11-MAO pathway may play an important role in stress-related brain disorders. Chronic stress is a risk factor for psychiatric illnesses, including depressive disorders, and is characterized by increased blood glucocorticoids and brain monoamine oxidase A (MAO A, which degrades monoamine neurotransmitters). This study elucidates the relationship between stress-induced MAO A and the transcription factor Kruppel-like factor 11 (KLF11, also called TIEG2, a member of the Sp/KLF- family), which inhibits cell growth. We report that 1) a glucocorticoid (dexamethasone) increases KLF11 mRNA and protein levels in cultured neuronal cells; 2) overexpressing KLF11 increases levels of MAO A mRNA and enzymatic activity, which is further enhanced by glucocorticoids; in contrast, siRNA-mediated KLF11 knockdown reduces glucocorticoid-induced MAO A expression in cultured neurons; 3) induction of KLF11 and translocation of KLF11 from the cytoplasm to the nucleus are key regulatory mechanisms leading to increased MAO A catalytic activity and mRNA levels because of direct activation of the MAO A promoter via Sp/KLF-binding sites; 4) KLF11 knockout mice show reduced MAO A mRNA and catalytic activity in the brain cortex compared with wild-type mice; and 5) exposure to chronic social defeat stress induces blood glucocorticoids and activates the KLF11 pathway in the rat brain, which results in increased MAO A mRNA and enzymatic activity. Thus, this study reveals for the first time that KLF11 is an MAO A regulator and is produced in response to neuronal stress, which transcriptionally activates MAO A. The novel glucocorticoid-KLF11-MAO A pathway may play a crucial role in modulating distinct pathophysiological steps in stress-related disorders.


Frontiers in Neuroscience | 2010

The New Inhibitor of Monoamine Oxidase, M30, has a Neuroprotective Effect Against Dexamethasone-Induced Brain Cell Apoptosis

Shakevia Johnson; Shawna Tazik; Deyin Lu; Chandra Johnson; Moussa B. H. Youdim; Junming Wang; Grazyna Rajkowska; Xiao-Ming Ou

Stress detrimentally affects the brain and body and can lead to or be accompanied by depression. Although stress and depression may contribute to each other, the exact molecular mechanism underlying the effects is unclear. However, there is a correlation between stress and an increase in glucocorticoid secretion which causes a subsequent increase in monoamine oxidase (MAO) activity during stress. Consequently, MAO inhibitors have been used as traditional antidepressant drugs. Cellular treatment with the synthetic glucocorticoid, dexamethasone (a cellular stressor), has been reported to markedly increase both MAO A and MAO B catalytic activities, as well as apoptosis. This study compares the neuroprotective abilities of M30 (a new generation inhibitor of both MAO A and MAO B) with rasagiline (Azilect®, another new MAO B inhibitor) and selegiline (Deprenyl®, a traditional MAO B inhibitor) in the prevention of dexamethasone-induced brain cell death and MAO activity in human neuroblastoma cells, SH-SY5Y. M30 demonstrated the highest inhibitory effect on MAO A; however, M30 showed the lowest inhibitory effect on MAO B enzymatic activity in comparison to rasagiline and selegiline. Although, M30 exhibited the greatest neuroprotective effect by decreasing cell death rates and apoptotic DNA damage compared to rasagiline and selegiline, these neuroprotective effects of M30 were, overall, similar to rasagiline. Summarily, M30 has a generally greater impact on neuroprotection than the MAO B inhibitors, selegiline and rasagiline. Our results suggest that M30 may have great potential in alleviating disorders involving increases in both MAO A and MAO B, such as stress-induced disorders.


Neurotoxicity Research | 2011

Ethanol Increases TIEG2–MAO B Cell Death Cascade in the Prefrontal Cortex of Ethanol-Preferring Rats

Xiao Ming Ou; Chandra Johnson; Deyin Lu; Shakevia Johnson; Ian A. Paul; Mark C. Austin; Abiye H. Iyo; Jose Javier Miguel-Hidalgo; Jia Luo; Richard L. Bell; Matthew Grunewald; Junming Wang; Donald B. Sittman

Brain cell loss has been reported in subjects with alcoholism. However, the molecular mechanisms are unclear. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and monoamine oxidase B (MAO B) reportedly play a role in cellular dysfunction with regards to ethanol exposure. We have recently reported that GAPDH protein expression was increased in the brains of rats fed with ethanol. Furthermore, GAPDH interacts with the transcriptional activator, transforming growth factor-beta-inducible early gene 2 (TIEG2), to augment TIEG2-mediated MAO B activation, resulting in neuronal cell damage due to ethanol exposure. The current study investigates whether the TIEG2–MAO B cascade is also active in the brains of rats fed with ethanol. Ten ethanol-preferring rats were fed with a liquid diet containing ethanol, with increasing amounts of ethanol up to a final concentration of 6.4% representing a final diet containing 36% of calories for 28xa0days. Ten control rats were fed the liquid diet without ethanol. The expression of TIEG2 protein, MAO B mRNA levels, MAO B catalytic activity, and the levels of anti-apoptotic protein Bcl 2 and apoptotic protein caspase 3 were determined in the prefrontal cortex of the rats. Ethanol significantly increased protein levels of TIEG2, active caspase 3, MAO B mRNA and enzyme activity, but significantly decreased Bcl 2 protein expression compared to control rats. In summary, ethanol increases the TIEG2–MAO B brain cell death cascade in rat brains, suggesting that the TIEG2–MAO B pathway is a novel pathway for brain cell damage resulting from ethanol exposure, and may contribute to chronic alcohol-induced brain damage.


Alcoholism: Clinical and Experimental Research | 2014

The Expression of KLF11 (TIEG2), a Monoamine Oxidase B Transcriptional Activator in the Prefrontal Cortex of Human Alcohol Dependence

Chinelo Udemgba; Shakevia Johnson; Craig A. Stockmeier; Jia Luo; Paul R. Albert; Junming Wang; Warren L. May; Grazyna Rajkowska; Sharonda Harris; Donald B. Sittman; Xiao-Ming Ou

BACKGROUNDnThe biochemical pathways underlying alcohol abuse and dependence are not well understood, although brain cell loss and neurotoxicity have been reported in subjects with alcohol dependence. Monoamine oxidase B (MAO B; an enzyme that catabolizes neurotransmitters such as dopamine) is consistently increased in this psychiatric illness. MAO B has been implicated in the pathogenesis of alcohol dependence and alcohol-induced brain neurotoxicity. Recently, the cell growth inhibitor protein, Kruppel-like factor 11 (KLF11), has been reported to be an MAO transcriptional activator. KLF11 is also known as TIEG2 (transforming growth factor-beta-inducible early gene 2) and mediates apoptotic cell death. This study investigates the protein expression of KLF11 and its relationship with MAO B using human postmortem prefrontal cortex from subjects with alcohol dependence.nnnMETHODSnTwelve subjects with alcohol dependence and the respective psychiatrically normal control subjects were investigated. Expression of KLF11 and MAO B proteins in the prefrontal cortex was measured by Western blot analysis. Correlation studies involving KLF11 and MAO B protein expression were performed. Localization of KLF11 in the human prefrontal cortex was also determined by immunohistochemistry.nnnRESULTSnLevels of KLF11 protein were significantly increased by 44% (p < 0.03) in the postmortem prefrontal cortex of subjects with alcohol dependence as compared to age- and gender-matched, psychiatrically normal control subjects. Furthermore, KLF11 levels were significantly and positively correlated with both the increased MAO B protein levels and blood alcohol content in alcohol-dependent subjects. In addition, KLF11 protein expression was visualized in both neuronal and glial cells.nnnCONCLUSIONSnThis novel study shows the important role of KLF11, an MAO transcriptional activator, in human alcohol dependence. It further supports that the KLF11-MAO B cell death cascade may contribute to chronic alcohol-induced brain damage. This argues a case for KLF11-MAO B inhibition as a novel therapeutic strategy that may impact this highly prevalent illness.


Frontiers in Aging Neuroscience | 2015

Differential contributions of ApoE4 and female sex to BACE1 activity and expression mediate Aβ deposition and learning and memory in mouse models of Alzheimer’s disease

Xu Hou; Samuel O. Adeosun; Qinli Zhang; Brett R. Barlow; Melissa Brents; Baoying Zheng; Junming Wang

Alzheimer’s disease (AD), the most common form of dementia, disproportionately affects women in both prevalence and severity. This increased vulnerability to AD in women is strongly associated with age-related ovarian hormone loss and apolipoprotein E 4 allele (ApoE4), the most important genetic risk factor for sporadic AD. Up to date, the mechanism involved in the interaction between ApoE4 and sex/gender in AD is still unclear. This study evaluated the sex-dependent ApoE4 effects on learning and memory, Aβ deposition and potential mechanisms, using mice bearing both sporadic (ApoE4) and familial (APPSwe, PS1M146V, tauP301L; 3xTg) AD risk factors and compared with sex- and age-matched 3xTg or nonTg mice. Compared to nonTg mice, transgenic mice of both sexes showed spatial learning and memory deficits in the radial arm water maze and novel arm discrimination tests at 20 months of age. However, at 10 months, only ApoE4/3xTg mice showed significant learning and memory impairment. Moreover, molecular studies of hippocampal tissue revealed significantly higher protein levels of Aβ species, β-site APP cleavage enzyme (BACE1) and Sp1, a transcription factor of BACE1, in female ApoE4/3xTg when compared with female nonTg, female 3xTg, and male ApoE4/3xTg mice. Significantly increased BACE1 enzymatic activities were observed in both male and female mice carrying ApoE4; however, only the females showed significant higher BACE1 expressions. Together, these data suggest that ApoE4 allele is associated with increased BACE1 enzymatic activity, while female sex plays an important role in increasing BACE1 expression. The combination of both provides a molecular basis for high Aβ pathology and the resultant hippocampus-dependent learning and memory deficits in female ApoE4 carriers.


Alcoholism: Clinical and Experimental Research | 2014

Diabetes‐Causing Gene, Kruppel‐Like Factor 11, Modulates the Antinociceptive Response of Chronic Ethanol Intake

Xiao Ming Ou; Chinelo Udemgba; Niping Wang; Xiaoli Dai; Gwen Lomberk; Seungmae Seo; Raul Urrutia; Junming Wang; Jeremy Duncan; Sharonda Harris; Carolyn A. Fairbanks; Xiao Zhang

BACKGROUNDnAlcohol (EtOH [ethanol]) is an antinociceptive agent, working in part, by reducing sensitivity to painful stimuli. The transcription factor Kruppel-like factor 11 (KLF11), a human diabetes-causing gene that also regulates the neurotransmitter metabolic enzymes monoamine oxidase (MAO), has recently been identified as an EtOH-inducible gene. However, its role in antinociception remains unknown. Consequently, we investigated the function of KLF11 in chronic EtOH-induced antinociception using a genetically engineered knockout mouse model.nnnMETHODSnWild-type (Klf11(+/+) ) and KLF11 knockout (Klf11(-/-) ) mice were fed a liquid diet containing EtOH for 28xa0days with increasing amounts of EtOH from 0% up to a final concentration of 6.4%, representing a final diet containing 36% of calories primarily from EtOH. Control mice from both genotypes were fed liquid diet without EtOH for 28xa0days. The EtOH-induced antinociceptive effect was determined using the tail-flick test before and after EtOH exposure (on day 29). In addition, the enzyme activity and mRNA levels of MAO A and MAO B were measured by real-time RT-PCR and enzyme assays, respectively.nnnRESULTSnEtOH produced an antinociceptive response to thermal pain in Klf11(+/+) mice, as expected. In contrast, deletion of KLF11 in the Klf11(-/-) mice abolished the EtOH-induced antinociceptive effect. The mRNA and protein levels of KLF11 were significantly increased in the brain prefrontal cortex of Klf11(+/+) mice exposed to EtOH compared with control Klf11(+/+) mice. Furthermore, MAO enzyme activities were affected differently in Klf11 wild-type versus Klf11 knockout mice exposed to chronic EtOH. Chronic EtOH intake significantly increased MAO B activity in Klf11(+/+) mice.nnnCONCLUSIONSnThe data show KLF11 modulation of EtOH-induced antinociception. The KLF11-targeted MAO B enzyme may contribute more significantly to EtOH-induced antinociception. Thus, this study revealed a new role for the KLF11 gene in the mechanisms underlying the antinociceptive effects of chronic EtOH exposure.


Alzheimers & Dementia | 2011

The impact of a neurogenic agent, allopregnanolone, in rodent models of Alzheimer's and Parkinson's disease

Junming Wang; Samuel O. Adeosun; Yun Jiao; Baoying Zheng; Sherry Henry; Xu Hou; Chenyou Sun; Amar K. Pani; Patrick B. Kyle; Jerry M. Farley; Ian A. Paul; Steven Bigler; Craig A. Stockmeier; Roberta Diaz Brinton; Richard J. Smeyne

ALLOPREGNANOLONE, IN RODENTMODELS OF ALZHEIMER’S AND PARKINSON’S DISEASE Junming Wang, Samuel Adeosun, Yun Jiao, Baoying Zheng, Sherry Henry, Xu Hou, Chenyou Sun, Amar Pani, Patrick Kyle, Jerry Farley, Ian Paul, Steven Bigler, Craig Stockmeier, Roberta Brinton, Richard Smeyne, University Mississippi Medical Center, Jackson, Mississippi, United States; University of Mississippi Medical Center, Jackson, Mississippi, United States; 3 St. Jude Children’s Hospital, Memphis, Tennessee, United States; 4 University of Southern California, Los Angeles, California, United States.


BioScience Trends | 2009

High throughput analysis of neural progenitor cell proliferation in adult rodent hippocampus.

Sherry Henry; Steven Bigler; Junming Wang


Alzheimers & Dementia | 2012

Motor deficits in mouse models of Alzheimer's disease

Samuel O. Adeosun; Xu Hou; Baoying Zheng; Ian A. Paul; Ronald W. Irwin; Roberta Diaz Brinton; Junming Wang

Collaboration


Dive into the Junming Wang's collaboration.

Top Co-Authors

Avatar

Shakevia Johnson

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Craig A. Stockmeier

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Donald B. Sittman

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ian A. Paul

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Roberta Diaz Brinton

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Baoying Zheng

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Deyin Lu

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mark C. Austin

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiao-Ming Ou

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Paul R. Albert

Ottawa Hospital Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge