Network


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

Hotspot


Dive into the research topics where Rihong Zhou is active.

Publication


Featured researches published by Rihong Zhou.


Journal of Cell Science | 2005

Phosphorylation of ezrin on threonine 567 produces a change in secretory phenotype and repolarizes the gastric parietal cell

Rihong Zhou; Lixin Zhu; Andrew Kodani; Paul Hauser; Xuebiao Yao; John G. Forte

Phosphorylation of the membrane-cytoskeleton linker protein ezrin has been functionally linked to acid secretion and vesicle recruitment to the apical secretory membrane in gastric parietal cells. Phosphorylation of the conserved T567 residue of ezrin has been shown to alter the N/C oligomerization of ezrin and promote the formation of actin-rich surface projections in other cells. To test the importance of T567 as a regulatory site for ezrin in parietal cell activation, we incorporated wild-type (WT) and mutant forms of ezrin, including the nonphosphorylatable T567A mutation and a mutant mimicking permanent phosphorylation, T567D. All ezrin constructs included C-terminal cyan-fluorescent protein (CFP) and were incorporated into adenoviral constructs for efficient introduction into cultured parietal cells from rabbit stomach. Fluorescence microscopy was used to localize CFP-ezrin and monitor morphological responses. Accumulation of a weak base (aminopyrine) was used to monitor receptor-mediated acid secretory response of the cultured cells. Similar to endogenous ezrin, WT and T567A CFP-ezrin localized heavily to apical membrane vacuoles with considerably lower levels associated with the surrounding basolateral membrane. Interestingly, H,K-ATPase within cytoplasmic tubulovesicles was incorporated into the apical vacuoles along with WT and T567A mutant ezrin. In these parietal cells secretagogue stimulation produced a striking vacuolar expansion associated with HCl secretion and the secretory phenotype. Expression of T567D CFP-ezrin was quite different, being rarely associated with apical vacuoles. T567D was more typically localized to the basolateral membrane, often associated with long spikes and fingerlike projections. Moreover, the cells did not display secretagogue-dependent morphological changes and, to our surprise, H,K-ATPase was recruited to the T567D CFP-ezrin-enriched basolateral projections. We conclude that T567 phosphorylation, which is probably regulated through Rho signaling pathway, may direct ezrin to membrane-cytoskeletal activity at the basolateral membrane and away from apical secretory activity. The large basolateral expansion is predicted to recruit membranes from sources not normally targeted to that surface.


Journal of Biological Chemistry | 2005

PALS1 specifies the localization of ezrin to the apical membrane of gastric parietal cells

Xinwang Cao; Xia Ding; Zhen Guo; Rihong Zhou; Fengsong Wang; Fei Long; Fang Wu; Feng Bi; Qichen Wang; Daiming Fan; John G. Forte; Maikun Teng; Xuebiao Yao

The ERM (ezrin/radixin/moesin) proteins provide a regulated linkage between membrane proteins and the cortical cytoskeleton and also participate in signal transduction pathways. Ezrin is localized to the apical membrane of parietal cells and couples the protein kinase A activation cascade to regulated HCl secretion in gastric parietal cells. Here, we show that the integrity of ezrin is essential for parietal cell activation and provide the first evidence that ezrin interacts with PALS1, an evolutionarily conserved PDZ and SH3 domain-containing protein. Our biochemical study verifies that ezrin binds to PALS1 via its N terminus and is co-localized with PALS1 to the apical membrane of gastric parietal cells. Furthermore, our study shows that PALS1 is essential for the apical localization of ezrin, as either suppression of PALS1 protein accumulation or deletion of the PALS1-binding domain of ezrin eliminated the apical localization of ezrin. Finally, our study demonstrates the essential role of ezrin-PALS1 interaction in the apical membrane remodeling associated with parietal cell secretion. Taken together, these results define a novel molecular mechanism linking ezrin to the conserved apical polarity complexes and their roles in polarized epithelial secretion of gastric parietal cells.


Molecular & Cellular Proteomics | 2006

Proteomic Identification and Functional Characterization of a Novel ARF6 GTPase-activating Protein, ACAP4

Zhiyou Fang; Yong Miao; Xia Ding; Hui Deng; Siqi Liu; Fengsong Wang; Rihong Zhou; Charles Watson; Chuanhai Fu; Qicong Hu; James W. Lillard; Michael Powell; Yong Chen; John G. Forte; Xuebiao Yao

ARF6 GTPase is a conserved regulator of membrane trafficking and actin-based cytoskeleton dynamics at the leading edge of migrating cells. A key determinant of ARF6 function is the lifetime of the GTP-bound active state, which is orchestrated by GTPase-activating protein (GAP) and GTP-GDP exchanging factor. However, very little is known about the molecular mechanisms underlying ARF6-mediated cell migration. To systematically analyze proteins that regulate ARF6 activity during cell migration, we performed a proteomic analysis of proteins selectively bound to active ARF6 using mass spectrometry and identified a novel ARF6-specific GAP, ACAP4. ACAP4 encodes 903 amino acids and contains two coiled coils, one pleckstrin homology domain, one GAP motif, and two ankyrin repeats. Our biochemical characterization demonstrated that ACAP4 has a phosphatidylinositol 4,5-bisphosphate-dependent GAP activity specific for ARF6. The co-localization of ACAP4 with ARF6 occurred in ruffling membranes formed upon AIF4 and epidermal growth factor stimulation. ACAP4 overexpression limited the recruitment of ARF6 to the membrane ruffles in the absence of epidermal growth factor stimulation. Expression of GTP hydrolysis-resistant ARF6Q67L resulted in accumulations of ACAP4 and ARF6 in the cytoplasmic membrane, suggesting that GTP hydrolysis is required for the ARF6-dependent membrane remodeling. Significantly the depletion of ACAP4 by small interfering RNA or inhibition of ARF6 GTP hydrolysis by overexpressing GAP-deficient ACAP4 suppressed ARF6-dependent cell migration in wound healing, demonstrating the importance of ACAP4 in cell migration. Thus, our study sheds new light on the biological function of ARF6-mediated cell migration.


American Journal of Physiology-cell Physiology | 2010

A possible mechanism for ezrin to establish epithelial cell polarity

Lixin Zhu; James M. Crothers; Rihong Zhou; John G. Forte

Ezrin is an important membrane/actin cytoskeleton linker protein, especially in epithelia. Ezrin has two important binding domains: an NH(2)-terminal region that binds to plasma membrane and a COOH-terminal region that binds to F-actin only after a conformational activation by phosphorylation at Thr567 of ezrin. The present experiments were undertaken to investigate the detailed cellular changes in the time course of expression of ezrin-T567 mutants (nonphosphorylatable T567A and permanent phospho-mimic T567D) in parietal cells and to assess ezrin distribution and its influence on the elaborate membrane recruitment processes of these cells. T567A mutant and wild-type (WT) ezrin were consistently localized to the apical plasma membrane, even with overexpression. On the other hand, T567D went first to apical membrane at early times and low expression levels, then accumulated mainly at the basal surface after 24 h. Overexpression of WT or T567A led to incorporation of internal membranes to apical vacuoles, while overexpression of T567D led to large incorporation of apical and intracellular membranes (including H-K-ATPase) to the basal surface. Differences in polar distribution of ezrin suggest a role for the linker protein in promoting formation and plasticity of membrane surface projections, forming the basis for a novel theory for ezrin as an organizer and regulator of membrane recruitment. A model simulating the cellular distribution of ezrin and its associated membrane- and F-actin-binding forms is given to predict redistributions observed with phosphorylation and mutant overexpression, and it can easily be modified as more specific information regarding binding constants and specific sites becomes available.


Journal of Biological Chemistry | 2003

Characterization of Protein Kinase A-mediated Phosphorylation of Ezrin in Gastric Parietal Cell Activation

Rihong Zhou; Xinwang Cao; Charles Watson; Yong Miao; Zhen Guo; John G. Forte; Xuebiao Yao


American Journal of Physiology-cell Physiology | 2007

High turnover of ezrin T567 phosphorylation: conformation, activity, and cellular function

Lixin Zhu; Rihong Zhou; Shelley Mettler; Tim Wu; Aennes Abbas; Joseph Delaney; John G. Forte


American Journal of Physiology-gastrointestinal and Liver Physiology | 2002

Syntaxin 3 is required for cAMP-induced acid secretion: streptolysin O-permeabilized gastric gland model.

David A. Ammar; Rihong Zhou; John G. Forte; Xuebiao Yao


Molecular Biology of the Cell | 2003

Polarized Distribution of IQGAP Proteins in Gastric Parietal Cells and Their Roles in Regulated Epithelial Cell Secretion

Rihong Zhou; Zhen Guo; Charles Watson; Emily Chen; Rong Kong; Wenxian Wang; Xuebiao Yao


American Journal of Physiology-cell Physiology | 2003

Myosin II is present in gastric parietal cells and required for lamellipodial dynamics associated with cell activation.

Rihong Zhou; Charles Watson; Chuanhai Fu; Xuebiao Yao; John G. Forte


The FASEB Journal | 2007

Reversible phosphorylation of ezrin T567: conformation, activity and physiology

Lixin Zhu; Rihong Zhou; Shelley Mettler; Tim Wu; Aennes Abbas; John G. Forte

Collaboration


Dive into the Rihong Zhou's collaboration.

Top Co-Authors

Avatar

John G. Forte

University of California

View shared research outputs
Top Co-Authors

Avatar

Xuebiao Yao

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Charles Watson

University of California

View shared research outputs
Top Co-Authors

Avatar

Lixin Zhu

University at Buffalo

View shared research outputs
Top Co-Authors

Avatar

Zhen Guo

Morehouse School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Chuanhai Fu

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Fengsong Wang

Anhui Medical University

View shared research outputs
Top Co-Authors

Avatar

Xia Ding

Beijing University of Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Xinwang Cao

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Yong Miao

University of Science and Technology of China

View shared research outputs
Researchain Logo
Decentralizing Knowledge