Network


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

Hotspot


Dive into the research topics where Dalong Ma is active.

Publication


Featured researches published by Dalong Ma.


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.


Autophagy | 2009

High-throughput functional screening for autophagy-related genes and identification of TM9SF1 as an autophagosome-inducing gene

Pengfei He; Zhi Peng; Ye Luo; Lan Wang; Peng Yu; Weiwei Deng; Yunqing An; Taiping Shi; Dalong Ma

Autophagy is a tightly regulated process responsible for the bulk degradation of most long-lived proteins and some organelles, which is associated with several forms of human disease including cancer, neurodegenerative disease, and cardiomyopathies. However, the molecular machinery involved in autophagy in mammalian cells remains poorly understood. Here, we describe a high-throughput, cell-based functional screening platform based on automated fluorescence microscopy system, that enable acquiring and quantitatively analyzing images of GFP-LC3 dots in cotransfected cells. From a library of 1,050 human cDNA clones, we identified three genes (TM9SF1, TMEM166, and TMEM74) whose overexpression induced high levels of autophagosome formation. In particular, overexpression of TM9SF1, which colocalized with LC3 according to the confocal assay, led to a significant increase in the number of GFP-LC3 dots. The results of transmission electron microscopy and immunoblotting to examine LC3-II levels further confirmed the ability of TM9SF1 to induce autophagy. Furthermore, knockdown of TM9SF1 expression by RNA interference could hamper starvation-induced autophagy. The functional screening platform therefore can be applied for high-throughput genomic screening candidate autophagy-related genes, which would provide new insights into underlying molecular mechanisms that may regulate autophagy in mammalian cells.


Apoptosis | 2007

TMEM166, a novel transmembrane protein, regulates cell autophagy and apoptosis

Lan Wang; Chuanfei Yu; Yang Lu; Pengfei He; Jinhai Guo; Chenying Zhang; Quansheng Song; Dalong Ma; Taiping Shi; Yingyu Chen

Programmed cell death can be divided into apoptosis and autophagic cell death. We describe the biological activities of TMEM166 (transmembrane protein 166, also known as FLJ13391), which is a novel lysosome and endoplasmic reticulum-associated membrane protein containing a putative TM domain. Overexpression of TMEM166 markedly inhibited colony formation in HeLa cells. Simultaneously, typical morphological characteristics consistent with autophagy were observed by transmission electron microscopy, including extensive autophagic vacuolization and enclosure of cell organelles by double-membrane structures. Further experiments confirmed that the overexpression of TMEM166 increased the punctate distribution of MDC staining and GFP-LC3 in HeLa cells, as well as the LC3-II/LC3-I proportion. On the other hand, TMEM166-transfected HeLa and 293T cells succumbed to cell death with hallmarks of apoptosis including phosphatidylserine externalization, loss of mitochondrial transmembrane potential, caspase activation and chromatin condensation. Kinetic analysis revealed that the appearance of autophagy-related biochemical parameters preceded the nuclear changes typical of apoptosis in TMEM166-transfected HeLa cells. Suppression of TMEM166 expression by small interference RNA inhibited starvation-induced autophagy in HeLa cells. These findings show for the first time that TMEM166 is a novel regulator involved in both autophagy and apoptosis.


Life Sciences | 2002

Transfer of anti-TFAR19 monoclonal antibody into HeLa cells by in situ electroporation can inhibit the apoptosis.

Min Rui; Yingyu Chen; Yingmei Zhang; Dalong Ma

Electroporation has been successfully used for the introduction of DNA, RNA, oligonucleotide and protein into eukaryotic and prokaryotic cells for the transformation and expression of various gene products. TFAR19 (TF-1 apoptosis-related gene 19), also designated PDCD5 (Programmed Cell Death 5), is cloned as an increased expression gene during the apoptotic process of TF-1 cell induced by cytokine withdrawal. It facilitates rather than induces apoptosis in different cell lines. To explore its molecular mechanism, we successfully transferred the anti-TFAR19 monoclonal antibody into HeLa cells by in situ electroporation and observed the apoptosis process of HeLa cells induced by etoposide with flow cytometry. We demonstrate that the introduction of anti-TFAR19 antibody can suppress the apoptosis accelerating effect of TFAR19 in its natural environment. This study shows that TFAR19 may be a critical factor for apoptosis; and transfer of monoclonal antibody into mammalian cells by in situ electroporation is a useful method to study the function of endogenous factors.


Molecular Cancer Research | 2009

Down-regulation of 3-phosphoinositide-dependent protein kinase-1 levels inhibits migration and experimental metastasis of human breast cancer cells.

Ying Liu; Jingna Wang; Min Wu; Wuzhou Wan; Ronghua Sun; De Yang; Xiangjun Sun; Dalong Ma; Guoguang Ying; Ning Zhang

High expression of 3-phosphoinositide–dependent protein kinase-1 (PDK1) has been detected in various invasive cancers. In the current study, we investigated its role in cancer cell migration and experimental metastasis. Down-regulation of PDK1 expression by small interference RNA markedly inhibited spontaneous migration and epidermal growth factor (EGF)–induced chemotaxis of human breast cancer cells. The defects were rescued by expressing wild-type PDK1. PDK1-depleted cells showed impaired EGF-induced actin polymerization and adhesion, probably due to a decrease in phosphorylation of LIM kinase/cofilin and integrin β1. Confocal microscopy revealed that EGF induced cotranslocation of PDK1 with Akt and protein kinase Cζ (PKCζ), regulators of LIM kinase, and integrin β1. Furthermore, PDK1 depletion dampened EGF-induced phosphorylation and translocation of Akt and PKCζ, suggesting that Akt and PKCζ functioned downstream of PDK1 in the chemotactic signaling pathway. In severe combined immunodeficiency mice, PDK1-depleted human breast cancer cells formed more slowly growing tumors and were defective in extravasation to mouse lungs after i.v. injection. Our results indicate that PDK1 plays an important role in regulating the malignant behavior of breast cancer cells, including their motility, through activation of Akt and PKCζ. Thus, PDK1, which increases its expression in cancer cells, can be used as a target for the development of novel therapies. (Mol Cancer Res 2009;7(6):944–54)


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.

Collaboration


Dive into the Dalong Ma'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

Taiping Shi

Chinese National Human Genome Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weiwei Deng

Chinese National Human Genome Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge