Network


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

Hotspot


Dive into the research topics where Wenling Han is active.

Publication


Featured researches published by Wenling Han.


Biochemical Journal | 2001

Molecular cloning and characterization of chemokine-like factor 1 (CKLF1), a novel human cytokine with unique structure and potential chemotactic activity

Wenling Han; Yaxin Lou; Junmin Tang; Yingmei Zhang; Yingyu Chen; Ying Li; Weifeng Gu; Jiaqiang Hueng; Liming Gui; Yan Tang; Feng Li; Quansheng Song; Chunhui Di; Lu Wang; Qun Shi; Ronghua Sun; Donglan Xia; Min Rui; Jian Tang; Dalong Ma

Cytokines are small proteins that have an essential role in the immune and inflammatory responses. The repertoire of cytokines is becoming diverse and expanding. Here we report the identification and characterization of a novel cytokine designated as chemokine-like factor 1 (CKLF1). The full-length cDNA of CKLF1 is 530 bp long and a single open reading frame encoding 99 amino acid residues. CKLF1 bears no significant similarity to any other known cytokine in its amino acid sequence. Expression of CKLF1 can be partly inhibited by interleukin 10 in PHA-stimulated U937 cells. Recombinant CKLF1 is a potent chemoattractant for neutrophils, monocytes and lymphocytes; moreover, it can stimulate the proliferation of murine skeletal muscle cells. These results suggest that CKLF1 might have important roles in inflammation and in the regeneration of skeletal muscle.


FEBS Letters | 2001

Nuclear translocation of PDCD5 (TFAR19): an early signal for apoptosis?

Yingyu Chen; Ronghua Sun; Wenling Han; Yingmei Zhang; Quansheng Song; Chunhui Di; Dalong Ma

The programmed cell death 5 (PDCD5) protein is a novel protein related to regulation of cell apoptosis. In this report, we demonstrate that the level of PDCD5 protein expressed in cells undergoing apoptosis is significantly increased compared with normal cells, then the protein translocates rapidly from the cytoplasm to the nucleus of cells. The appearance of PDCD5 in the nuclei of apoptotic cells precedes the externalization of phosphatidylserine and fragmentation of chromosome DNA. This phenomenon is parallel to the loss of mitochondrial membrane potential, independent of the feature of apoptosis‐inducing stimuli and also independent of the cell types and the apoptosis modality. In conclusion, the nuclear translocation of PDCD5 is a universal earlier event of the apoptotic process, and may be a novel early marker for apoptosis.


Genomics | 2003

Identification of eight genes encoding chemokine-like factor superfamily members 1-8 (CKLFSF1-8) by in silico cloning and experimental validation.

Wenling Han; Peiguo Ding; Mingxu Xu; Lu Wang; Min Rui; Shuang Shi; Yanan Liu; Ying Zheng; Yingyu Chen; Tian Yang; Dalong Ma

TM4SF11 is only 102 kb from the chemokine gene cluster composed of SCYA22, SCYD1, and SCYA17 on chromosome 16q13. CKLF maps on chromosome 16q22. CKLFs have some characteristics associated with the CCL22/MDC, CX3CL1/fractalkine, CCL17/TARC, and TM4SF proteins. Bioinformatics based on CKLF2 cDNA and protein sequences in combination with experimental validation identified eight novel genes designated chemokine-like factor superfamily members 1-8 (CKLFSF1-8). CKLFSF1-8 form gene clusters; the sequence identities between CKLF2 and CKLFSF1-8 are from 12.5 to 39.7%. Most of the CKLFSFs have alternative RNA splicing forms. CKLFSF1 has a CC motif and higher sequence similarity with chemokines than with any of the other CKLFSFs. CKLFSF8 shares 39.3% amino acid identity with TM4SF11. CKLFSF1 links the CKLFSF family with chemokines, and CKLFSF8 links it with TM4SF. The characteristics of CKLFSF2-7 are intermediate between CKLFSF1 and CKLFSF8. This indicates that CKLFSF represents a novel gene family between the SCY and the TM4SF gene families.


Cancer Research | 2009

CMTM3, Located at the Critical Tumor Suppressor Locus 16q22.1, Is Silenced by CpG Methylation in Carcinomas and Inhibits Tumor Cell Growth through Inducing Apoptosis

Yu Wang; Jisheng Li; Yan Cui; Ting Li; Ka Man Ng; Hua Geng; Henan Li; Xingsheng Shu; Hongyu Li; Wei Liu; Bing Luo; Qian Zhang; Tony Mok; Wei Zheng; Xiaoyan Qiu; Gopesh Srivastava; Jun Yu; Joseph J.Y. Sung; Anthony T.C. Chan; Dalong Ma; Qian Tao; Wenling Han

Closely located at the tumor suppressor locus 16q22.1, CKLF-like MARVEL transmembrane domain-containing member 3 and 4 (CMTM3 and CMTM4) encode two CMTM family proteins, which link chemokines and the transmembrane-4 superfamily. In contrast to the broad expression of both CMTM3 and CMTM4 in normal human adult tissues, only CMTM3 is silenced or down-regulated in common carcinoma (gastric, breast, nasopharyngeal, esophageal, and colon) cell lines and primary tumors. CMTM3 methylation was not detected in normal epithelial cell lines and tissues, with weak methylation present in only 5 of 35 (14%) gastric cancer adjacent normal tissues. Furthermore, immunohistochemistry showed that CMTM3 protein was absent in 12 of 35 (34%) gastric and 1 of 2 colorectal tumors, which was well correlated with its methylation status. The silencing of CMTM3 is due to aberrant promoter CpG methylation that could be reversed by pharmacologic demethylation. Ectopic expression of CMTM3 strongly suppressed the colony formation of carcinoma cell lines. In addition, CMTM3 inhibited tumor cell growth and induced apoptosis with caspase-3 activation. Thus, CMTM3 exerts tumor-suppressive functions in tumor cells, with frequent epigenetic inactivation by promoter CpG methylation in common carcinomas.


Life Sciences | 2006

Chemokine-like factor 1 is a functional ligand for CC chemokine receptor 4 (CCR4)

Ying Wang; Yingmei Zhang; Xue Yang; Wenling Han; Yanan Liu; Qianmei Xu; Rui Zhao; Chunhui Di; Quansheng Song; Dalong Ma

Abstract Chemokine-like factor 1 (CKLF1) exhibits chemotactic effects on leukocytes. Its amino acid sequence shares similarity with those of TARC/CCL17 and MDC/CCL22, the cognate ligands for CCR4. The chemotactic effects of CKLF1 for CCR4-transfected cells could be desensitized by TARC/CCL17 and markedly inhibited by PTX. CKLF1 induced a calcium flux in CCR4-transfected cells and fully desensitized a subsequent response to TARC/CCL17, and TARC/CCL17 could partly desensitize the response to CKLF1. CKLF1 caused significant receptor internalization in pCCR4-EGFP transfected cells. Taken together, CKLF1 is a novel functional ligand for CCR4.


Clinical Cancer Research | 2007

CMTM5 Exhibits Tumor Suppressor Activities and Is Frequently Silenced by Methylation in Carcinoma Cell Lines

Luning Shao; Yan Cui; Hongyu Li; Yanan Liu; Hongshan Zhao; Yu Wang; Yingmei Zhang; Ka Man Ng; Wenling Han; Dalong Ma; Qian Tao

Purpose:CMTM5 (CKLF-like MARVEL transmembrane domain containing member 5) is located at 14q11.2, a locus associated with multiple cancers. It has six RNA splicing variants with CMTM5-v1 as the major one. We explored its expression pattern in normal tissues and tumor cell lines, as well as its functions in carcinoma cells. Experimental Design: We evaluated CMTM5 expression by semiquantitative reverse transcription-PCR (RT-PCR) in normal tissues and carcinoma cell lines of cervical, breast, nasopharyngeal, lung, hepatocellular, esophageal, gastric, colon, and prostate. We further examined CMTM5 promoter methylation in these cell lines. We also analyzed CMTM5 expression after 5-aza-2′-deoxycytidine treatment and genetic demethylation and the functional consequences of restoring CMTM5 in HeLa and PC-3 cells. Results: CMTM5-v1 is broadly expressed in human normal adult and fetal tissues, but undetectable or down-regulated in most carcinoma cell lines. Its promoter methylation was detected in virtually all the silenced or down-regulated cell lines. The silencing of CMTM5 could be reversed by pharmacologic demethylation or genetic double-knockout of DNMT1 and DNMT3B, indicating methylation-mediated mechanism. Restoration of CMTM5-v1 suppressed carcinoma cell proliferation, migration, and invasion. Conclusions: These results indicate that CMTM5 exhibits tumor suppressor activities, but with frequent epigenetic inactivation in carcinoma cell lines.


Cellular Signalling | 2008

PTPIP51, a novel 14–3–3 binding protein, regulates cell morphology and motility via Raf–ERK pathway

Chuanfei Yu; Wenling Han; Taiping Shi; Bingfeng Lv; Qihua He; Yanfei Zhang; Ting Li; Yingmei Zhang; Quansheng Song; Lu Wang; Dalong Ma

Cell migration plays a critical role during the development of most organisms and the process of malignant tumor metastasis. In the present study, we investigated the role of PTPIP51 (protein tyrosine phosphatase interacting protein 51) in cell motility. Overexpression of PTPIP51 induced cell elongation, increased cell migration, adhesion, and spreading, while downregulation of PTPIP51 had the opposite effects. We demonstrated here, that PTPIP51 could regulate ERK activity on Raf level, since MEK inhibitor and dominant-negative Raf-1 but not Ras could inhibit the ERK activation induced by PTPIP51. Further studies proved that PTPIP51 could interact with Raf-1 through 14-3-3, suggesting that PTPIP51 is a regulator of the Raf-MEK-ERK cascade through modulation of Raf-1 by 14-3-3. In addition, two redundant 14-3-3 binding domains in the PTPIP51 protein have been identified by deletion/mutation studies. We conclude that PTPIP51 regulates cell morphology and cell motility via interaction with Raf-1 through 14-3-3, and that PTPIP51 binds to 14-3-3 through two redundant binding domains.


Oncogene | 2014

A novel 3p22.3 gene CMTM7 represses oncogenic EGFR signaling and inhibits cancer cell growth

Henan Li; Jisheng Li; Y. Su; Yichao Fan; Xiaohuan Guo; Lili Li; Xianwei Su; Rong Rong; Jianming Ying; Xiaoning Mo; K. Liu; Z. Zhang; F. Yang; G. Jiang; Jun Wang; Yingmei Zhang; Defu Ma; Qian Tao; Wenling Han

Deletion of 3p12-22 is frequent in multiple cancer types, indicating the presence of critical tumor-suppressor genes (TSGs) at this region. We studied a novel candidate TSG, CMTM7, located at the 3p22.3 CMTM-gene cluster, for its tumor-suppressive functions and related mechanisms. The three CMTM genes, CMTM6, 7 and 8, are broadly expressed in human normal adult tissues and normal epithelial cell lines. Only CMTM7 is frequently silenced or downregulated in esophageal and nasopharyngeal cell lines, but uncommon in other carcinoma cell lines. Immunostaining of tissue microarrays for CMTM7 protein showed its downregulation or absence in esophageal, gastric, pancreatic, liver, lung and cervix tumor tissues. Promoter CpG methylation and loss of heterozygosity were both found contributing to CMTM7 downregulation. Ectopic expression of CMTM7 in carcinoma cells inhibits cell proliferation, motility and tumor formation in nude mice, but not in immortalized normal cells, suggesting a tumor inhibitory role of CMTM7. The tumor-suppressive function of CMTM7 is associated with its role in G1/S cell cycle arrest, through upregulating p27 and downregulating cyclin-dependent kinase 2 (CDK2) and 6 (CDK6). Moreover, CMTM7 could promote epidermal growth factor receptor (EGFR) internalization, and further suppress AKT signaling pathway. Thus, our findings suggest that CMTM7 is a novel 3p22 tumor suppressor regulating G1/S transition and EGFR/AKT signaling during tumor pathogenesis.


Biochemical and Biophysical Research Communications | 2008

CMTM3 can affect the transcription activity of androgen receptor and inhibit the expression level of PSA in LNCaP cells.

Yu Wang; Ting Li; Xiaoyan Qiu; Xiaoning Mo; Yingmei Zhang; Quansheng Song; Dalong Ma; Wenling Han

CMTM is a novel family of proteins linking chemokines and TM4SF. Several members of this family are highly expressed in testes and regulate androgen receptor (AR) transcription activity. One member of this family, CMTM3, has the highest expression level in testes and contains one leucine zipper and two LXXLL motifs. As assessed with the dual-luciferase reporter assay, overexpression of CMTM3 represses AR transactivation, while knocking down it can increase AR transactivation. Moreover, CMTM3 inhibits prostate-specific antigen (PSA) expression in LNCaP cells at both mRNA and protein levels with no obvious influence on AR expression. Taken together, CMTM3 may play some roles in the maturation and maintenance of the male reproduction.


Journal of Cellular Physiology | 2007

CMTM8 induces caspase-dependent and -independent apoptosis through a mitochondria-mediated pathway

Caining Jin; Ying Wang; Wenling Han; Yingmei Zhang; Qihua He; Dan Li; Caihua Yin; Linjie Tian; Dazhen Liu; Quanshen Song; Dalong Ma

The mitochondria‐mediated apoptotic pathway is regulated by members of the Bcl‐2 family. Epidermal growth factor (EGF) induces Bad phosphorylation at Ser112 via mitogen‐activated protein kinase (MAPK), impairing its binding to Bcl‐2 and Bcl‐xL and interfering with their anti‐apoptotic functions. In the current study, we utilized Western blot, immunofluorescence, flow cytometry, and confocal microscopy to examine the effects of CMTM8 overexpression on apoptosis. Our data indicated levels of Bad‐S112 phosphorylation were lower in CMTM8‐transfected cells compared to pCDB‐transfected cells. Caspase‐dependent and independent mediated apoptosis, induced by CMTM8 overexpression, was facilitated by the mitochondria and inhibited by knockdown of Bad or overexpression of Bcl‐xL. Previous research in our laboratory also demonstrated CMTM8 attenuated EGFR‐mediated signaling pathways by decreasing ERK1/2phosphorylation levels. These data implicate CMTM8 as a negative regulator of EGF‐induced signaling, with potential use as a novel therapeutic gene for EGFR‐targeted anticancer gene therapy. J. Cell. Physiol. 211: 112–120, 2007.

Collaboration


Dive into the Wenling Han's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge