UB Vice President for Research and Economic Development markup
Jian Feng UB Profile
Physiology and Biophysics
Professor
549 Biomedical Research Building
Buffalo NY 14214
716-829-2345
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Jian Feng
PubMedId: 32972199
1: Experimental biology and medicine (Maywood, N.J.), vol. 246, 2020
Authors: Bo Li, Houbo Jiang, Hong Li, Boyang Zhang, Malcolm Slaughter, Zhen Yan, Jian Feng
PubMedId: 33070653
1: Experimental biology and medicine (Maywood, N.J.), vol. 246, 2020
Authors: Lin Lin, Aiyi Liu, Hanqin Li, Jian Feng, Zhen Yan
Journal-article: Journal of Alzheimer's Disease, vol. 70, pp. 1175-1185, 2019
Authors: Zhixing Hu, Hanqin Li, Houbo Jiang, Yong Ren, Xinyang Yu, Jingxin Qiu, Aimee B Stablewski, Boyang Zhang, Michael J Buck, Jian Feng
PubMedId: 32426495
1: Science advances, vol. 6, 2019
Authors: Hanqin Li, Zhixing Hu, Houbo Jiang, Jiali Pu, Ilana Selli, Jingxin Qiu, Baorong Zhang, Jian Feng
PubMedId: 32587369
1: Scientific reports, vol. 10, 2019
Authors: Hanqin Li, Houbo Jiang, Xinzhen Yin, Jonathan E Bard, Baorong Zhang, Jian Feng
PubMedId: 31255287
1: Biochemical and biophysical research communications, vol. 516, 2019
Authors: Hong Li, Houbo Jiang, Boyang Zhang, Jian Feng
PubMedId: 30149462
1: Journal of Parkinson's disease, vol. 8, 2018
Authors: Zhimin Xu, Xingkun Chu, Houbo Jiang, Haley Schilling, Shengdi Chen, Jian Feng
PubMedId: 28110217
1: Redox biology, vol. 11, 2017
Authors: Ping Zhong, Zhixing Hu, Houbo Jiang, Zhen Yan, Jian Feng
PubMedId: 28467897
Journal-article: Cell Reports, vol. 19, pp. 1033-1044, 2017
Authors: Jian Feng
PubMedId: 26900955
1: Cell cycle (Georgetown, Tex.), vol. 15, 2016
Authors: Jiali Pu, David Frescas, Baorong Zhang, Jian Feng
PubMedId: 25956682
1: Experimental biology and medicine (Maywood, N.J.), vol. 240, 2015
Authors: Zhixing Hu, Jiali Pu, Houbo Jiang, Ping Zhong, Jingxin Qiu, Feng Li, Xiaomin Wang, Baorong Zhang, Zhen Yan, Jian Feng
PubMedId: 26218671
1: Stem cells and development, vol. 24, 2015
Authors: Houbo Jiang, Zhimin Xu, Ping Zhong, Yong Ren, Gaoyang Liang, Haley A. Schilling, Zihua Hu, Yi Zhang, Xiaomin Wang, Shengdi Chen, Zhen Yan, Jian Feng
PubMedId: 26639555
Journal-article: Nature Communications, vol. 6, 2015
Authors: Yong Ren, Houbo Jiang, Zhixing Hu, Kevin Fan, Jun Wang, Stephen Janoschka, Xiaomin Wang, Shaoyu Ge, Jian Feng
Keywords: Dopamine Induced pluripotent stem cells Microtubule Parkin Parkinson's disease
Abstract: Parkinson's disease (PD) is characterized by the degeneration of nigral dopaminergic (DA) neurons and non-DA neurons in many parts of the brain. Mutations of parkin, an E3 ubiquitin ligase that strongly binds to microtubules, are the most frequent cause of recessively inherited PD. The lack of robust PD phenotype in parkin knockout mice suggests a unique vulnerability of human neurons to parkin mutations. Here, we show that the complexity of neuronal processes as measured by total neurite length, number of terminals, number of branch points, and Sholl analysis was greatly reduced in induced pluripotent stem cell (iPSC)-derived TH(+) or TH(-) neurons from PD patients with parkin mutations. Consistent with these, microtubule stability was significantly decreased by parkin mutations in iPSC-derived neurons. Overexpression of parkin, but not its PD-linked mutant nor green fluorescent protein, restored the complexity of neuronal processes and the stability of microtubules. Consistent with these, the microtubule-depolymerizing agent colchicine mimicked the effect of parkin mutations by decreasing neurite length and complexity in control neurons while the microtubule-stabilizing drug taxol mimicked the effect of parkin overexpression by enhancing the morphology of parkin-deficient neurons. The results suggest that parkin maintains the morphological complexity of human neurons by stabilizing microtubules.
PubMedId: 25332110
Journal-article: Stem cells (Dayton, Ohio), vol. 33, 2014
Authors: Houbo Jiang, Yong Ren, Eunice Y Yuen, Ping Zhong, Mahboobe Ghaedi, Zhixing Hu, Gissou Azabdaftari, Kazuhiro Nakaso, Zhen Yan, Jian Feng
Abstract: Parkinson's disease (PD) is defined by the degeneration of nigral dopaminergic (DA) neurons and can be caused by monogenic mutations of genes such as parkin. The lack of phenotype in parkin knockout mice suggests that human nigral DA neurons have unique vulnerabilities. Here we generate induced pluripotent stem cells from normal subjects and PD patients with parkin mutations. We demonstrate that loss of parkin in human midbrain DA neurons greatly increases the transcription of monoamine oxidases and oxidative stress, significantly reduces DA uptake and increases spontaneous DA release. Lentiviral expression of parkin, but not its PD-linked mutant, rescues these phenotypes. The results suggest that parkin controls dopamine utilization in human midbrain DA neurons by enhancing the precision of DA neurotransmission and suppressing dopamine oxidation. Thus, the study provides novel targets and a physiologically relevant screening platform for disease-modifying therapies of PD.
PubMedId: 22314364
Journal-article: Nature communications, vol. 3, 2011
Authors: Wenhua Liu, Fei Dou, Jian Feng, Zhen Yan
Journal-article: Neurobiology of Aging, vol. 32, pp. 1818-1826, 2011
Authors: Houbo Jiang, Dongmei Cheng, Wenhua Liu, Junmin Peng, Jian Feng
Journal-article: Biochemical and Biophysical Research Communications, vol. 395, pp. 471-476, 2010
Authors: Yong Ren, Houbo Jiang, Dingyuan Ma, Kazuhiro Nakaso, Jian Feng
Abstract: Parkin, whose mutations cause Parkinson disease (PD), controls oxidative stress by limiting the expression of monoamine oxidases (MAO)--mitochondrial enzymes responsible for the oxidative de-amination of dopamine. Here, we show that parkin performed this function by increasing the ubiquitination and degradation of estrogen-related receptors (ERR), orphan nuclear receptors that play critical roles in the transcription regulation of many nuclear-encoded mitochondrial proteins. All three ERRs (, and ) increased the transcription of MAOs A and B; the effects were abolished by parkin, but not by its PD-linked mutants. Parkin bound to ERRs and increased their ubiquitination and degradation. In fibroblasts from PD patients with parkin mutations or brain slices from parkin knockout mice, degradation of ERRs was significantly attenuated. The results reveal the molecular mechanism by which parkin suppresses the transcription of MAOs to control oxidative stress induced by dopamine oxidation.
PubMedId: 21177257
Journal-article: Human molecular genetics, vol. 20, 2010
Authors: Jerome A. Roth, Steven Singleton, Jian Feng, Michael Garrick, Prasad N. Paradkar
Abstract: Abnormal iron accumulation is linked to a variety of neurological disorders and may contribute to the progressive damage seen in these diseases. The biochemical processes responsible for iron accumulation are not known but are likely to entail alteration in transport into injured brain areas. The major transport protein responsible for uptake of iron is divalent metal transporter 1 (DMT1) and recent studies demonstrate that the 1B species is regulated post-translationally by degradation via the proteasomal pathway. As reported in this paper, the E3 ligase, parkin, when over-expressed in SH-SY5Y cells, results in a decrease in 1B-DMT1 isoforms and also a significant reduction in manganese transport and toxicity. Incubating cells over-expressing parkin with the proteasomal inhibitor, MG-132, restores 1B-DMT1 levels emphasizing that the observed changes are caused by degradation via the proteasomal pathway. Expression of the 1B species of DMT1 was also shown to be elevated in human lymphocytes containing a homozygous deletion of exon 4 of parkin and in brains of parkin knockout animals. Immunoprecipitation and immunofluorescent studies confirm that parkin co-localizes with DMT1 in SH-SY5Y cells transfected with wild-type parkin. These results demonstrate that parkin is the E3 ligase responsible for ubiquitination of the 1B species of DMT1.
PubMedId: 20089134
Journal-article: Journal of Neurochemistry, vol. 113, pp. 454-464, 2010
Authors: E Y Yuen, W Liu, I N Karatsoreos, Y Ren, J Feng, B S McEwen, Z Yan
Journal-article: Molecular Psychiatry, vol. 16, pp. 156-170, 2010
Authors: B. Porton, R. M. Rodriguiz, L. E. Phillips, J. W. Gilbert, J. Feng, P. Greengard, H.-T. Kao, W. C. Wetsel
Journal-article: Genes, Brain and Behavior, vol. 9, pp. 257-268, 2010
Authors: Eunice Y Yuen, Wenhua Liu, Ilia N Karatsoreos, Jian Feng, Bruce S McEwen, Zhen Yan
Abstract: The prefrontal cortex (PFC), a key brain region controlling cognition and emotion, is strongly influenced by stress. While chronic stress often produces detrimental effects on these measures, acute stress has been shown to enhance learning and memory, predominantly through the action of corticosteroid stress hormones. We used a combination of electrophysiological, biochemical, and behavioral approaches in an effort to identify the cellular targets of acute stress. We found that behavioral stressors in vivo cause a long-lasting potentiation of NMDAR- and AMPAR-mediated synaptic currents via glucocorticoid receptors (GRs) selectively in PFC pyramidal neurons. This effect is accompanied by increased surface expression of NMDAR and AMPAR subunits in acutely stressed animals. Furthermore, behavioral tests indicate that working memory, a key function relying on recurrent excitation within networks of PFC neurons, is enhanced by acute stress via a GR-dependent mechanism. These results have identified a form of long-term potentiation of synaptic transmission induced by natural stimuli in vivo, providing a potential molecular and cellular mechanism for the beneficial effects of acute stress on cognitive processes subserved by PFC.
PubMedId: 19666502
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 106, 2009
Authors: Guojun Chen, Paul Chen, Huibing Tan, Da Ma, Fei Dou, Jian Feng, Zhen Yan
Journal-article: Neurobiology of Aging, vol. 29, pp. 1795-1804, 2008
Authors: Hung-Teh Kao, Ping Li, Helen M. Chao, Stephen Janoschka, Kara Pham, Jian Feng, Bruce S. Mcewen, Paul Greengard, Vincent A. Pieribone, Barbara Porton
PubMedId: 18271024
Journal-article: The Journal of Comparative Neurology, vol. 507, pp. 1860-1870, 2008
Authors: Michael M Madden, Wenjiao Song, Paul G Martell, Yong Ren, Jian Feng, Qing Lin
Abstract: Protein ubiquitination is a widespread protein posttranslational modification in eukaryotes that regulates essentially every aspect of cellular processes. The attachment of ubiquitin to a protein substrate is accomplished through an enzymatic cascade involving the actions of an activating enzyme (E1), a conjugating enzyme (E2), and a ligase (E3). There are more than 600 E3 ligases estimated to exist in the human genome that regulate the targeting specificity of protein ubiquitination. To understand the dynamic role of protein ubiquitination in biological processes, robust tools need to be developed which can be employed to establish the substrate specificity of each of these E3 ligases. In this report, we show that the ubiquitin carboxyl-terminally derived peptide probes can serve as modest ubiquitin surrogates for the ubiquitination pathway. In the E1-catalyzed probe adenylation assay, peptide probe 3 with a RLRGG recognition sequence exhibited the highest activity, with the k cat/ K 1/2 determined to be 1.1 x 10 (4) M (-1) s (-1), roughly 470-fold lower than that of ubiquitin. The rate of transfer from the E1 peptide probe thioesters to E2 showed clear sequence dependency, with peptide probe 4 with an LRLRGG recognition sequence showed the fastest rate ( t 1/2 = 0.9 min), essentially identical to that of ubiquitin ( t 1/2 = 0.8 min) under our assay conditions. Furthermore, peptide probes 4 and 8 also exhibited the selective, parkin-mediated labeling of tubulins in a semipurified tubulin-parkin complex. Finally, these carboxyl-terminally derived peptide probes were shown to label the ubiquitination substrates in fraction II of the rabbit reticulocyte lysate with an efficiency parallel to their substrate properties. The selective use of these ubiquitin carboxyl-terminally derived peptide probes by the ubiquitination pathway suggests that perhaps more potent peptide ubiquitination probes based on the ubiquitin C-terminal scaffold can be developed through additional structural optimization.
PubMedId: 18307316
Journal-article: Biochemistry, vol. 47, 2008
Authors: Qian Jiang, Yong Ren, Jian Feng
Abstract: Histone deacetylase 6 (HDAC6), a microtubule-associated tubulin deacetylase, plays a significant role in the formation of protein aggregates in many neurodegenerative disorders. Parkin, a protein-ubiquitin E3 ligase linked to Parkinson's disease, accumulates at the centrosome in a microtubule-dependent manner in response to proteasome inhibition. Here, we show that the centrosome recruitment of parkin was mediated by its direct binding to HDAC6 through multiple interaction domains. The tubulin deacetylase activity of HDAC6 was required for the accumulation of parkin as well as its dispersion upon the reversal of proteasome inhibition. The bidirectional movements of parkin required intact microtubule network and were dependent on dynein and kinesin 1, respectively. Tubulin deacetylation increases microtubule dynamicity and may thus facilitate microtubule-based trafficking of the parkin-HDAC6 complex. The results suggest that HDAC6 acts as a sensor of proteasome inhibition and directs the trafficking of parkin by using different motor proteins.
PubMedId: 19036992
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 28, 2008
Authors: Yong Ren, Houbo Jiang, Fang Yang, Kazuhiro Nakaso, Jian Feng
Abstract: Mitogen-activated protein kinases, originally known as microtubule-associated protein (MAP) kinases, are activated in response to a variety of stimuli. Here we report that microtubule-depolymerizing agents such as colchicine or nocodazole induced strong activation of MAP kinases including JNK, ERK, and p38. This effect was markedly attenuated by parkin, whose mutations are linked to Parkinson disease (PD). Our previous study has shown that parkin stabilizes microtubules through strong interactions mediated by three independent domains. We found that each of the three microtubule-binding domains of parkin was sufficient to reduce MAP kinase activation induced by microtubule depolymerization. The ability to attenuate microtubule depolymerization and the ensuing MAP kinase activation was abrogated in B-lymphocytes and fibroblasts derived from PD patients with parkin mutations such as exon 4 deletion. Such mutations produced truncated parkin proteins lacking any microtubule binding domain and prevented parkin from protecting midbrain dopaminergic neurons against microtubule-depolymerizing toxins such as rotenone or colchicine. Consistent with these, blocking MAP kinase activation in midbrain dopaminergic neurons by knocking down MAP kinase kinases (MKK) significantly reduced the selective toxicity of rotenone or colchicine. Conversely, overexpression of MAP kinases caused marked toxicities that were significantly attenuated by parkin. Thus, the results suggest that parkin protects midbrain dopaminergic neurons against microtubule-depolymerizing PD toxins such as rotenone by stabilizing microtubules to attenuate MAP kinase activation.
PubMedId: 19074146
Journal-article: The Journal of biological chemistry, vol. 284, 2008
Authors: Yong Ren, Jian Feng
Abstract: As a major co-morbidity of Parkinson's disease (PD), depression is associated with the loss of serotonergic neurons. Our recent study has shown that midbrain dopaminergic neurons are particularly vulnerable to microtubule-depolymerizing agents including rotenone, an environmental toxin linked to PD. Here we show that rotenone also selectively killed serotonergic neurons in midbrain neuronal cultures. Its selective toxicity was significantly decreased by the microtubule-stabilizing drug taxol and mimicked by microtubule-depolymerizing agents such as colchicine and nocodazole. Microtubule depolymerization induced by rotenone or colchicine caused vesicle accumulation in the soma and killed serotonergic neurons through a mechanism dependent on serotonin metabolism in the cytosol. Blocking serotonin synthesis or degradation, as well as application of antioxidants, significantly reduced the selective toxicity of rotenone or colchicine. Inhibition of vesicular sequestration of serotonin exerted a selective toxicity on serotonergic neurons that was mitigated by blocking serotonin metabolism. Over-expression of parkin, a protein-ubiquitin E3 ligase that strongly binds to microtubules, greatly attenuated the selective toxicity of rotenone or colchicine. The protective effects of parkin were abrogated by its PD-linked mutations. Together, our results suggest that rotenone and parkin affect the survival of serotonergic neurons by impacting on microtubules in opposing manners.
PubMedId: 17587308
Journal-article: Journal of neurochemistry, vol. 103, 2007
Authors: Jian Feng
Abstract: Parkinson's disease (PD) is characterized by the selective loss of nigral dopaminergic (DA) neurons, which have long axons enriched with microtubules. Depolymerization of microtubules by PD toxins such as rotenone disrupts vesicular transport. The ensuing accumulation of vesicles in the cell body leads to increased cytosolic concentration of dopamine due to leakage of the vesicles. Elevated oxidative stress induced by dopamine oxidation may thus trigger the selective demise of DA neurons. Many strategies have been developed to protect DA neurons by stabilizing microtubules either directly or through intracellular signaling cascades. On the other hand, parkin, one of the most frequently mutated genes in PD, encodes for a protein-ubiquitin E3 ligase that strongly binds to microtubules. Parkin stabilizes microtubules through three domains that provide strong and independent interactions with tubulin and microtubules. These interactions anchor parkin on microtubules and may facilitate its E3 ligase activity on misfolded proteins transported along microtubules. Thus, parkin and rotenone, two prominent genetic and environmental factors linked to PD, act in an opposing manner on the same molecular target in the cell, microtubules, whose destruction underlies the selective vulnerability of dopaminergic neurons.
PubMedId: 17079513
Journal-article: The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry, vol. 12, 2006
Authors: Houbo Jiang, Qian Jiang, Wenhua Liu, Jian Feng
Abstract: Mutations of parkin are linked to early onset Parkinson disease. Here we show that stable transfection of parkin in the human dopaminergic neuroblastoma cell line SH-SY5Y markedly reduced the activities of both monoamine oxidase (MAO) A and B. The amount of 3,4-dihydroxyphenylacetic acid, which is produced during dopamine oxidation by MAO, was greatly reduced by parkin overexpression. Radioligand binding assays showed that MAO binding sites were decreased accordingly. Consistent with these, MAO-B protein level was much lower, whereas the amount of MAO-A protein was not determined due to the lack of a suitable antibody. Co-transfection of either MAO with parkin in HEK293 cells did not significantly alter ubiquitination and degradation of each MAO. When we measured MAO expression by real-time quantitative reverse transcription-PCR, marked reductions were seen in SH-SY5Y cells stably expressing parkin compared with the parental cells or a control line stably transfected with luciferase. In addition, parkin mutants defective in E3 ligase activity exhibited different effects on MAO expression. We found that parkin also significantly decreased mRNA levels of both MAOs in the mouse fibroblast cell line NIH3T3. Furthermore, MAO expression was significantly increased in human B lymphocyte cell lines derived from Parkinson disease patients with homozygous but not heterozygous deletion of exon 4 of parkin. Together these results suggest that parkin suppresses MAO expression. This function may limit the production of reactive oxygen species generated by MAO in dopamine oxidation and would, thus, be beneficial to the survival of dopaminergic neurons.
PubMedId: 16455660
Journal-article: The Journal of biological chemistry, vol. 281, 2006
Authors: Qian Jiang, Zhen Yan, Jian Feng
Abstract: Parkinson disease is characterized by the selective degeneration of dopaminergic (DA) neurons in substantia nigra. Long term epidemiological studies have implicated exposure to agricultural pesticides as a significant risk factor. Systemic administration of rotenone, a widely used pesticide, causes selective degeneration of nigral DA neurons and Parkinson disease-like symptoms in rats. Our previous study has shown that the microtubule depolymerizing activity of rotenone plays a critical role in its selective toxicity on DA neurons. Rotenone toxicity is mimicked by the microtubule-depolymerizing drug colchicine and attenuated by the microtubule-stabilizing agent taxol. Here we show that nerve growth factor (NGF) significantly reduced rotenone toxicity on TH(+) neurons in midbrain neuronal cultures. The protective effect of NGF was completely abolished by inhibiting the microtubule-associated protein kinase kinase (MEK) and partially reversed by blocking phosphatidylinositol 3-kinase. In addition, NGF decreased colchicine toxicity on TH(+) neurons in a manner dependent on MEK but not phosphatidylinositol 3-kinase. The protective effect of NGF against rotenone toxicity was occluded by the microtubule-stabilizing drug taxol. In a MEK-dependent manner, NGF significantly attenuated rotenone- or colchicine-induced microtubule depolymerization and ensuing accumulation of vesicles in the soma and elevation in protein carbonyls. Moreover, other neurotrophic factors such as brain-derived neurotrophic factor and glia cell line-derived neurotrophic factor also reduced rotenone- or colchicine-induced microtubule depolymerization and death of TH(+) through a MEK-dependent mechanism. Thus, our results suggest that neurotrophic factors activate the microtubule-associated protein kinase pathway to stabilize microtubules, and this action significantly attenuates rotenone toxicity on dopaminergic neurons.
PubMedId: 16887804
Journal-article: The Journal of biological chemistry, vol. 281, 2006
Authors: Fang Yang, Qian Jiang, Jinghui Zhao, Yong Ren, Mark D Sutton, Jian Feng
Abstract: Mutations of parkin, a protein-ubiquitin isopeptide ligase (E3), appear to be the most frequent cause of familial Parkinson's disease (PD). Our previous studies have demonstrated that parkin binds strongly to alpha/beta tubulin heterodimers and microtubules. Here we show that the strong binding between parkin and tubulin, as well as that between parkin and microtubules, was mediated by three independent domains: linker, RING1, and RING2. These redundant strong interactions made it virtually impossible to separate parkin from microtubules by high concentrations of salt (3.8 m) or urea (0.5 m). Parkin co-purified with tubulin and was found in highly purified tubulin preparation. Expression of either full-length parkin or any of its three microtubule-binding domains significantly attenuated colchicine-induced microtubule depolymerization. The abilities of parkin to bind to and stabilize microtubules were not affected by PD-linked mutations that abrogate its E3 ligase activity. Thus, the tubulin/microtubule-binding activity of parkin and its E3 ligase activity are independent. The strong binding between parkin and tubulin/microtubules through three redundant interaction domains may not only stabilize microtubules but also guarantee the anchorage of this E3 ligase on microtubules. Because many misfolded proteins are transported on microtubules, the localization of parkin on microtubules may provide an important environment for its E3 ligase activity toward misfolded substrates.
PubMedId: 15737990
Journal-article: The Journal of biological chemistry, vol. 280, 2005
Authors: Houbo Jiang, Qian Jiang, Jian Feng
Abstract: Mutations of parkin, a protein-ubiquitin E3 ligase, are linked to Parkinson's disease (PD). Although a variety of parkin substrates have been identified, none of these is selectively expressed in dopaminergic neurons, whose degeneration plays a critical role in PD. Here we show that parkin significantly increased dopamine uptake in the human dopaminergic neuroblastoma cell line SH-SY5Y. This effect was accompanied by increased V(max) of dopamine uptake and unchanged K(m). Consistent with this, increased binding sites for dopamine transporter (DAT) ligand were observed in SH-SY5Y cells overexpressing parkin. The results were confirmed when parkin was transfected in HEK293 cells stably expressing DAT. In these cells, parkin enhanced the ubiquitination and degradation of DAT, increased its cell surface expression, and augmented dopamine uptake. The effects of parkin were significantly abrogated by its PD-causing mutations. Because the cell surface expression of functional DAT requires its oligomerization, misfolded DAT, induced either by the protein glycosylation inhibitor tunicamycin or by its C-terminal truncation, significantly attenuated cell surface expression of native DAT and reduced dopamine uptake. Expression of parkin, but not its T240R mutant, significantly alleviated these detrimental effects of misfolded DAT. Thus, our studies suggest that parkin increases dopamine uptake by enhancing the ubiquitination and degradation of misfolded DAT, so as to prevent it from interfering with the oligomerization and cell surface expression of native DAT. This function of parkin would enhance the precision of dopaminergic transmission, increase the efficiency of dopamine utilization, and reduce dopamine toxicity on neighboring cells.
PubMedId: 15492001
Journal-article: The Journal of biological chemistry, vol. 279, 2004
Authors: Jinghui Zhao, Yong Ren, Qian Jiang, Jian Feng
Abstract: Parkin is a protein-ubiquitin E3 ligase linked to Parkinson's disease. Although several substrates of parkin have been identified, the subcellular location for parkin to recognize and ubiquitinate its targets is unclear. Here we report that parkin was accumulated in the centrosome when SH-SY5Y or transfected HEK293 cells were treated with the proteasome inhibitor lactacystin. The specific recruitment of parkin was dependent on concentration and duration of the treatment, and was accompanied by the centrosomal accumulation of ubiquitinated proteins and CDCrel-1, a substrate of parkin. The recruitment of parkin was apparently mediated through its binding to gamma-tubulin, which has been shown to accumulate in the centrosome in response to misfolded proteins. Furthermore, the effect was abrogated by the microtubule-depolymerizing drug colchicine or the microtubule-stabilizing drug taxol, which indicates that the intact microtubule network is required for the centrosomal recruitment of parkin. Taken together, our data suggest that the lactacystin-induced accumulation of parkin in the centrosome plays a significant role in the ubiquitination of misfolded substrates accumulated there. This process may provide a subcellular locale for parkin to ubiquitinate and degrade protein aggregates critically involved in the pathogenesis of Parkinson's disease.
PubMedId: 12928331
Journal-article: Journal of cell science, vol. 116, 2003
Authors: Yong Ren, Jinghui Zhao, Jian Feng
Journal-article: The Journal of Neuroscience, vol. 23, pp. 3316-3324, 2003
Authors: Xiao-Hong Ma, Ping Zhong, Zhenglin Gu, Jian Feng, Zhen Yan
Journal-article: The Journal of Neuroscience, vol. 23, pp. 1159-1168, 2003
Authors: Xiang Cai, Jorge Flores-Hernandez, Jian Feng, Zhen Yan
Journal-article: The Journal of Physiology, vol. 540, pp. 743-759, 2002
Authors: Xiang Cai, Jorge Flores-Hernandez, Jian Feng, Zhen Yan
Journal-article: The Journal of Physiology, vol. 540, pp. 743-759, 2002
Authors: Jian Feng, Ping Chi, Thomas A. Blanpied, Yimei Xu, Ana Maria Magarinos, Adriana Ferreira, Reisuke H. Takahashi, Hung-Teh Kao, Bruce S. McEwen, Timothy A. Ryan, George J. Augustine, Paul Greengard
Journal-article: The Journal of Neuroscience, vol. 22, pp. 4372-4380, 2002
Authors: N. Carpino, R. Kobayashi, H. Zang, Y. Takahashi, S.-T. Jou, J. Feng, H. Nakajima, J. N. Ihle
Journal-article: Molecular and Cellular Biology, vol. 22, pp. 7491-7500, 2002
Authors: Vincent A. Pieribone, Barbara Porton, Beatrice Rendon, Jian Feng, Paul Greengard, Hung-Teh Kao
Journal-article: The Journal of Comparative Neurology, vol. 454, pp. 105-114, 2002
Authors: Jian Feng, Xiang Cai, Jinghui Zhao, Zhen Yan
Journal-article: The Journal of Neuroscience, vol. 21, pp. 6502-6511, 2001
Authors: Adriana Ferreira, Hung-Teh Kao, Jian Feng, Mark Rapoport, Paul Greengard
Journal-article: The Journal of Neuroscience, vol. 20, pp. 3736-3744, 2000
Authors: Demin Wang, Jian Feng, Renren Wen, Jean-Christophe Marine, Mark Y. Sangster, Evan Parganas, Angelika Hoffmeyer, Carl W. Jackson, John L. Cleveland, Peter J. Murray, James N. Ihle
Journal-article: Immunity, vol. 13, pp. 25-35, 2000
Authors: J. Feng, Z. Yan, A. Ferreira, K. Tomizawa, J. A. Liauw, M. Zhuo, P. B. Allen, C. C. Ouimet, P. Greengard
Journal-article: Proceedings of the National Academy of Sciences, vol. 97, pp. 9287-9292, 2000
Authors: Zhen Yan, Linda Hsieh???Wilson, Jian Feng, Kazuhito Tomizawa, Patrick B. Allen, Allen A. Fienberg, Angus C. Nairn, Paul Greengard
Journal-article: Nature Neuroscience, vol. 2, pp. 13-17, 1999
Authors: H.-T. Kao, B. Porton, A. J. Czernik, J. Feng, G. Yiu, M. Haring, F. Benfenati, P. Greengard
Journal-article: Proceedings of the National Academy of Sciences, vol. 95, pp. 4667-4672, 1998
Authors: F. W. Quelle, J. Wang, J. Feng, D. Wang, J. L. Cleveland, J. N. Ihle, G. P. Zambetti
Journal-article: Genes & Development, vol. 12, pp. 1099-1107, 1998
Authors: JAMES N. IHLE, WILLIAM THIERFELDER, STEPHAN TEGLUND, DIMITRIOS STRAVAPODIS, DEMIN WANG, JIAN FENG, EVAN PARGANAS
Journal-article: Annals of the New York Academy of Sciences, vol. 865, pp. 1-9, 1998
Authors: F Kohlhuber, N C Rogers, D Watling, J Feng, D Guschin, J Briscoe, B A Witthuhn, S V Kotenko, S Pestka, G R Stark, J N Ihle, I M Kerr
Journal-article: Molecular and Cellular Biology, vol. 17, pp. 695-706, 1997
Authors: J Feng, B A Witthuhn, T Matsuda, F Kohlhuber, I M Kerr, J N Ihle
Journal-article: Molecular and Cellular Biology, vol. 17, pp. 2497-2501, 1997
Presented by CUBS © 2024