Network


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

Hotspot


Dive into the research topics where Jinbin Zhai is active.

Publication


Featured researches published by Jinbin Zhai.


Journal of Cell Biology | 2008

PyK2 and FAK connections to p190Rho guanine nucleotide exchange factor regulate RhoA activity, focal adhesion formation, and cell motility

Yangmi Lim; Ssang-Taek Lim; Alok Tomar; Margaret L. Gardel; Joie A. Bernard-Trifilo; Xiao Lei Chen; Sean Uryu; Rafaela Cañete-Soler; Jinbin Zhai; Hong Lin; William W. Schlaepfer; Perihan Nalbant; Gary M. Bokoch; Dusko Ilic; Clare M. Waterman-Storer; David D. Schlaepfer

Integrin binding to matrix proteins such as fibronectin (FN) leads to formation of focal adhesion (FA) cellular contact sites that regulate migration. RhoA GTPases facilitate FA formation, yet FA-associated RhoA-specific guanine nucleotide exchange factors (GEFs) remain unknown. Here, we show that proline-rich kinase-2 (Pyk2) levels increase upon loss of focal adhesion kinase (FAK) in mouse embryonic fibroblasts (MEFs). Additionally, we demonstrate that Pyk2 facilitates deregulated RhoA activation, elevated FA formation, and enhanced cell proliferation by promoting p190RhoGEF expression. In normal MEFs, p190RhoGEF knockdown inhibits FN-associated RhoA activation, FA formation, and cell migration. Knockdown of p190RhoGEF-related GEFH1 does not affect FA formation in FAK−/− or normal MEFs. p190RhoGEF overexpression enhances RhoA activation and FA formation in MEFs dependent on FAK binding and associated with p190RhoGEF FA recruitment and tyrosine phosphorylation. These studies elucidate a compensatory function for Pyk2 upon FAK loss and identify the FAK–p190RhoGEF complex as an important integrin-proximal regulator of FA formation during FN-stimulated cell motility.


The Journal of Neuroscience | 2005

Aldolases A and C Are Ribonucleolytic Components of a Neuronal Complex That Regulates the Stability of the Light-Neurofilament mRNA

Rafaela Cañete-Soler; Konda S. Reddy; Dean R. Tolan; Jinbin Zhai

A 68 nucleotide segment of the light neurofilament (NF-L) mRNA, spanning the translation termination signal, participates in regulating the stability of the transcript in vivo. Aldolases A and C, but not B, interact specifically with this segment of the transcript in vitro. Aldolases A and C are glycolytic enzymes expressed in neural cells, and their mRNA binding activity represents a novel function of these isozymes. This unsuspected new activity was first uncovered by Northwestern blotting of a brainstem/spinal cord cDNA library. It was confirmed by two-dimensional fractionation of mouse brain cytosol followed by Northwestern hybridization and protein sequencing. Both neuronal aldolases interact specifically with the NF-L but not the heavy neurofilament mRNA, and their binding to the transcript excludes the poly(A)-binding protein (PABP) from the complex. Constitutive ectopic expression of aldolases A and C accelerates the decay of a neurofilament transgene (NF-L) driven by a tetracycline inducible system. In contrast, mutant transgenes lacking mRNA sequence for aldolase binding are stabilized. Our findings strongly suggest that aldolases A and C are regulatory components of a light neurofilament mRNA complex that modulates the stability of NF-L mRNA. This modulation likely involves endonucleolytic cleavage and a competing interaction with the PABP. Interactions of aldolases A and C in NF-L expression may be linked to regulatory pathways that maintain the highly asymmetrical form and function of large neurons.


The Journal of Neuroscience | 2004

3′ Untranslated Region in a Light Neurofilament (NF-L) mRNA Triggers Aggregation of NF-L and Mutant Superoxide Dismutase 1 Proteins in Neuronal Cells

Hong Lin; Jinbin Zhai; Rafaela Cañete-Soler; William W. Schlaepfer

The pathogenesis of neurodegenerative diseases is believed to involve abnormal aggregation of proteins, but the mechanisms initiating protein aggregation are unclear. Here we report a novel phenomenon that could be instrumental in triggering protein aggregation in neurodegenerative diseases. We show that the 3′ untranslated region (3′UTR) of a light neurofilament (NF-L) transcript enhances the reactivity of its own translated product and leads to loss of solubility and aggregation of NF-L protein and to coaggregation of mutant superoxide dismutase 1 (SOD1) protein. Full-length mouse NF-L cDNAs, with and without NF-L 3′UTR, were fused to the C terminus of a green fluorescent protein (GFP) reporter gene, and the GFP-tagged NF-L proteins were examined in transfected Neuro2a cells. The GFP-tagged NF-L protein expressed from the transgene containing NF-L 3′UTR, but not from the transgene lacking NF-L 3′UTR, colocalizes with endogenous heavy neurofilament protein and, at high-level expression, leads to loss of solubility and aggregation of GFP-tagged NF-L protein. Aggregation of GFP-tagged NF-L protein triggers coaggregation and loss of solubility of coexpressed DsRed-tagged mutant (G93A) SOD1 protein but not wild-type SOD1 protein. Deletional mutagenesis maps the RNA sequence causing aggregation of GFP-tagged NF-L protein to the proximal 45 nucleotides of NF-L 3′UTR. This is the site of a major destabilizing element in NF-L RNA and binding site for RNA-binding proteins. Our findings support a working model whereby NF-L RNA, or cognate RNA-binding factors, enhances the reactivity of NF-L protein and provides a triggering mechanism leading to aggregation of NF-L and other proteins in neurodegenerative diseases.


Molecular Brain Research | 2003

Cytoplasmic retention sites in p190RhoGEF confer anti-apoptotic activity to an EGFP-tagged protein

Junhua Wu; Jinbin Zhai; Hong Lin; Zhenying Nie; Wei Wen Ge; Laura García-Bermejo; Ruth J. Muschel; William W. Schlaepfer; Rafaela Cañete-Soler

p190RhoGEF is a large multi-functional protein with guanine nucleotide exchange (GEF) activity. The C-terminal region of p190RhoGEF is a highly interactive domain that binds multiple factors, including proteins with anti-apoptotic activities. We now report that transfection of EGFP-tagged p190RhoGEF protects Neuro 2a cells from stress-induced apoptosis and that anti-apoptotic activity is localized to cytoplasmic retention sequences (CRS-1 and CRS-2) in the C-terminal region of p190RhoGEF. Cytoplasmic retention is conferred to an EGFP fluorescent marker when fused to either CRS-1 or CRS-2. Both cytoplasmic retention and anti-apoptotic activity are lost by deleting CRS-1 and CRS-2 in the p190RhoGEF sequence and can be recovered by restoring either CRS-1 or CRS-2 to the EGFP-tagged sequence. Since the CRS-1 and CRS-2 contain the JIP-1 and 14-3-3 binding sites, we propose that anti-apoptotic activity may be conferred by the binding of p190RhoGEF to JIP-1 or 14-3-3, possibly by altering their interactive properties or nucleocytoplasmic movements. Taken together, our findings support a model whereby multiple interactions of p190RhoGEF confer homeostatic properties to differentiated neurons and may link neuronal homeostasis to the regulation of NF-L expression.


Journal of Biological Chemistry | 2003

Direct Interaction of Focal Adhesion Kinase with p190RhoGEF

Jinbin Zhai; Hong Lin; Zhenying Nie; Junhua Wu; Rafaela Cañete-Soler; William W. Schlaepfer; David D. Schlaepfer


Human Molecular Genetics | 2007

Disruption of neurofilament network with aggregation of light neurofilament protein: a common pathway leading to motor neuron degeneration due to Charcot-Marie-Tooth disease-linked mutations in NFL and HSPB1

Jinbin Zhai; Hong Lin; Jean-Pierre Julien; William W. Schlaepfer


Journal of Biological Chemistry | 2001

Identification of a Novel Interaction of 14-3-3 with p190RhoGEF

Jinbin Zhai; Hong Lin; Mohammad Shamim; William W. Schlaepfer; Rafaela Cañete-Soler


Journal of Biological Chemistry | 2001

p190RhoGEF Binds to a Destabilizing Element in the 3′ Untranslated Region of Light Neurofilament Subunit mRNA and Alters the Stability of the Transcript

Rafaela Cañete-Soler; Junhua Wu; Jinbin Zhai; Mohammad Shamim; William W. Schlaepfer


Human Molecular Genetics | 2005

RNA-binding protein is involved in aggregation of light neurofilament protein and is implicated in the pathogenesis of motor neuron degeneration

Hong Lin; Jinbin Zhai; William W. Schlaepfer


The Journal of Neuroscience | 2002

Untranslated Element in Neurofilament mRNA Has Neuropathic Effect on Motor Neurons of Transgenic Mice

Zhenying Nie; Junhua Wu; Jinbin Zhai; Hong Lin; Weiwen Ge; William W. Schlaepfer; Rafaela Cañete-Soler

Collaboration


Dive into the Jinbin Zhai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Lin

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Junhua Wu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mohammad Shamim

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Weiwen Ge

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Alok Tomar

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge