Network


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

Hotspot


Dive into the research topics where Xiuli Peng is active.

Publication


Featured researches published by Xiuli Peng.


Toxicon | 2016

Mechanism of ESAT-6 membrane interaction and its roles in pathogenesis of Mycobacterium tuberculosis

Xiuli Peng; Jianjun Sun

The 6-kDa early secreted antigenic target (ESAT-6; EsxA) of Mycobacterium tuberculosis was first identified as a potent T-cell antigen, and it is now recognized as a pore-forming toxin that is essential for virulence of M.xa0tuberculosis. ESAT-6 is secreted through the ESX-1 secretion system (Type VII) of M.xa0tuberculosis and has been implicated to mediate mycobacterial cytosolic translocation within the host macrophages by rupturing the phagosomal membranes. Recent studies have made significant progresses in understanding of the mechanism of ESAT-6 membrane interaction and its role in M.xa0tuberculosis pathogenesis, but important questions still remain to be answered. Here, we summarize the current progress in study of ESAT-6 membrane interaction and its roles in pathogenesis and discuss some of the key remaining questions for future investigation.


Developmental and Comparative Immunology | 2016

Roles of Toll-like receptors 2 and 6 in the inflammatory response to Mycoplasma gallisepticum infection in DF-1 cells and in chicken embryos.

Wei Tian; Chengcheng Zhao; Qingchuang Hu; Jianjun Sun; Xiuli Peng

While Mycoplasma gallisepticum (MG) is a major pathogen that causes chronic respiratory diseases in chicken, the molecular mechanism of MG infection is not clear. In this study, we investigated the roles of Toll-like receptor 2 (TLR2) and 6 (TLR6) in MG infection. We found that TLR2 type 2 (TLR2-2) and TLR6 had differential expressions in chicken embryo fibroblasts (DF-1xa0cells), where TLR6 was highly expressed, but TLR2-2 was barely expressed. Upon MG infection, TLR6 expression was upregulated, followed by upregulation of downstream factors, MyD88, NF-κB, IL2, IL6, and TNF-α. Knockdown of TLR6 expression by shRNA abolished the MG-induced inflammatory responses. More interestingly, in the presence of TLR6, TLR2-2 didnt respond to MG infection in DF-1xa0cells. When TLR6 was knocked down by shRNA, however, TLR2 was upregulated upon MG infection, which was followed by upregulation of proinflammatory genes. Finally, we tested effects of the MG infection on expression of TLR2-2 and TLR6 in the lungs and trachea tissues of chicken embryos. We found both TLR2-2 and TLR6 were upregulated upon MG infection, followed by upregulation of the downstream NF-κB-mediated inflammatory responses. This study was the first to report the differential roles of TLR2-2 and TLR6 in MG-infected DF-1xa0cells and chicken embryos.


FEBS Letters | 2016

Characterization of differential pore‐forming activities of ESAT‐6 proteins from Mycobacterium tuberculosis and Mycobacterium smegmatis

Xiuli Peng; Guozhong Jiang; Wei Liu; Qi Zhang; Wei Qian; Jianjun Sun

Mycobacterium tuberculosis ESAT‐6 (MtbESAT‐6) plays essential roles in pathogenesis. MtbESAT‐6 exhibits a unique pore‐forming activity (PFA) that is not found in its ortholog from non‐pathogenic Mycobacterium smegmatis (MsESAT‐6). Here, we characterized the differential PFAs and found that exchange of I25‐H26/T25‐A26 between two proteins reciprocally affected their PFAs. MtbESAT‐6(IH/TA) had ~ 40% reduction, while MsESAT‐6(TA/IH) fully acquired its activity similar to MtbESAT‐6. Mutations of A17E, K38T, N67L or R74Q on MtbESAT‐6(IH/TA) further reduced the activity, with MtbESAT‐6(IH/TA‐17) being the lowest. This study suggests I25‐H26 as the pH‐sensor essential for MsESAT‐6 to fully acquire the activity, while multiple residues contributed to MtbESAT‐6 PFA.


International Journal of Molecular Sciences | 2015

Gga-miR-101-3p Plays a Key Role in Mycoplasma gallisepticum (HS Strain) Infection of Chicken.

Jiao Chen; Zaiwei Wang; Dingren Bi; Yue Hou; Yabo Zhao; Jianjun Sun; Xiuli Peng

Mycoplasma gallisepticum (MG), one of the most pathogenic Mycoplasma, has caused tremendous economic loss in the poultry industry. Recently, increasing evidence has suggested that micro ribonucleic acids (miRNAs) are involved in microbial pathogenesis. However, little is known about potential roles of miRNAs in MG infection of chicken. In the present study, using miRNA Solexa sequencing we have found that gga-miR-101-3p was up-regulated in the lungs of MG-infected chicken embryos. Moreover, gga-miR-101-3p regulated expression of the host enhancer of zeste homolog 2 (EZH2) through binding to the 3’ un-translated region (3’-UTR) of EZH2 gene. Over-expression of gga-miR-101-3p significantly inhibited EZH2 expression and hence inhibited proliferation of chicken embryonic fibroblast (DF-1 cells) by blocking the G1-to-S phase transition. Similar results were obtained in MG-infected chicken embryos and DF-1 cells, where gga-miR-101-3p was significantly up-regulated, while EZH2 was significantly down-regulated. This study reveals that gga-miR-101-3p plays an important role in MG infection through regulation of EZH2 expression and provides a new insight into the mechanisms of MG pathogenesis.


Comparative Biochemistry and Physiology Part D: Genomics and Proteomics | 2017

Identification of differentially expressed miRNAs through high-throughput sequencing in the chicken lung in response to Mycoplasma gallisepticum HS

Yabo Zhao; Yue Hou; Kang Zhang; Bo Yuan; Xiuli Peng

Mycoplasma gallisepticum (MG) infects chickens, causes chronic respiratory diseases (CRD) and severely damages the poultry industry. It has been suggested that micro-ribonucleic acids (miRNAs) are involved in microbial pathogenesis. Here, we identified miRNAs that are associated with MG infection in chicken lungs at 3 and 10days post-infection by deep sequencing. Thirty-six down-regulated and 9 up-regulated miRNAs belonging to 31 miRNA families were detected at 3days post-infection, whereas 50 down-regulated and 18 up-regulated miRNAs belonging to 41 miRNA families were found at 10days post-infection. The 45 and 68 differentially expressed miRNAs at 3 and 10days target 6280 and 7181 genes, respectively. In this study, 8 candidate novel chicken miRNAs were identified. Analyses via GO, KEGG, miRNA-GO-network, path-net and gene-net showed that these altered miRNAs might be involved in regulating the host response to MG infection by targeting genes in many pathways, such as the MAPK pathway, focal adhesion, Wnt pathway, endocytosis, Jak/STAT pathway, phosphatidylinositol pathway, adherens junctions, regulation of actin cytoskeleton among others. These analyses indicate that the MAPK pathway may be a key regulatory route. Also, the miR-8 family, miR-499 family, miR-17 family, and PIK3 family genes, as well as the MAP2K1 and RAC1 genes, might be important in MG infection. miR-20 of the miR-17 family was further confirmed by RT-qPCR. The important miRNAs, mRNAs and pathways associated with MG infection in chicken are valuable for further research. Our data provide new insights into the mechanism of these miRNAs on the regulation of host-MG interactions.


PLOS ONE | 2015

Endothelin Receptor B2 (EDNRB2) Gene Is Associated with Spot Plumage Pattern in Domestic Ducks (Anas platyrhynchos).

Ling Li; Dan Li; Li Liu; Shijun Li; Yanping Feng; Xiuli Peng; Yanzhang Gong

Endothelin receptor B subtype 2 (EDNRB2) is a seven-transmembrane G-protein coupled receptor. In this study, we investigated EDNRB2 gene as a candidate gene for duck spot plumage pattern according to studies of chicken and Japanese quail. The entire coding region was cloned by the reverse transcription polymerase chain reaction (RT-PCR). Sequence analysis showed that duck EDNRB2 cDNA contained a 1311bp open reading frame and encoded a putative protein of 436 amino acids residues. The transcript shared 89%-90% identity with the counterparts in other avian species. A phylogenetic tree based on amino acid sequences showed that duck EDNRB2 was evolutionary conserved in avian clade. The entire coding region of EDNRB2 were sequenced in 20 spot and 20 non-spot ducks, and 13 SNPs were identified. Two of them (c.940G>A and c.995G>A) were non-synonymous substitutions, and were genotyped in 647 ducks representing non-spot and spot phenotypes. The c.995G>A mutation, which results in the amino acid substitution of Arg332His, was completely associated with the spot phenotype: all 152 spot ducks were carriers of the AA genotype and the other 495 individuals with non-spot phenotype were carriers of GA or GG genotype, respectively. Segregation in 17 GA×GG and 22 GA×GA testing combinations confirmed this association since the segregation ratios and genotypes of the offspring were in agreement with the hypothesis. In order to investigate the underlying mechanism of the spot phenotype, MITF gene was used as cell type marker of melanocyte progenitor cells while TYR and TYRP1 gene were used as cell type markers of mature melanocytes. Transcripts of MITF, TYR and TYRP1 gene with expected size were identified in all pigmented skin tissues while PCR products were not obtained from non-pigmented skin tissues. It was inferred that melanocytes are absent in non-pigmented skin tissues of spot ducks.


Applied Microbiology and Biotechnology | 2016

Mycoplasma gallisepticum (HS strain) surface lipoprotein pMGA interacts with host apolipoprotein A-I during infection in chicken.

Fuli Hu; Chengcheng Zhao; Dingren Bi; Wei Tian; Jiao Chen; Jianjun Sun; Xiuli Peng

The adhesin protein from Mycoplasma gallisepticum (HS strain), namely pMGA1.2, is required for M. gallisepticum (MG) infection in chicken. However, the host factor(s) that interact with pMGA1.2 is not known. In this study, we prepared the membrane fraction of trachea epithelial cells from chicken embryos. Using an improved virus overlay protein blot assay (VOPBA) and glutathione S-transferase (GST) pull-down assay, we found that pMGA1.2 specifically bound to a ∼30xa0kDa host protein. This host protein was further identified by mass spectrometry as chicken apolipoprotein A-I (ApoA-I). We expressed and purified the recombinant ApoA-I protein in Escherichia coli and confirmed that it bound to the purified pMGA1.2 protein in vitro. Transiently expressed pMGA1.2 and ApoA-I were colocalized in HeLa cells. Finally, we designed small interfering RNA (siRNA) molecules to knock down the expression of either ApoA-I or pMGA1.2, which inhibited the MG-induced cell cycle disruption in cells of chicken embryo fibroblast cell line (DF-1). Similarly, knockdown of ApoA-I inhibited the cilia loss and damage in chicken trachea cells in MG infection. In summary, ApoA-I may be an essential host factor in MG infection through interacting with pMGA1.2.


Gene | 2017

gga-miR-99a targets SMARCA5 to regulate Mycoplasma gallisepticum (HS strain) infection by depressing cell proliferation in chicken

Yabo Zhao; Zaiwei Wang; Yue Hou; Kang Zhang; Xiuli Peng

Mycoplasma gallisepticum (MG), one of the primary etiological agents of poultry chronic respiratory disease, has caused significant economic losses worldwide, and increasing evidence has recently indicated that miRNAs are involved in its microbial pathogenesis. gga-miR-99a, a member of the miR-99 family, plays an essential role in a variety of diseases. Through miRNA Solexa sequencing, we previously found that gga-miR-99a is significantly down-regulated in the lungs of MG-infected chicken embryos. In this study, we further verified that the expression of gga-miR-99 was significantly down-regulated in both MG-infected lungs and a chicken embryonic fibroblast cell line (DF-1) by qPCR. Moreover, we predicted and validated SMARCA5 as its target gene through a luciferase reporter assay, qPCR, and western blot analysis. The over-expression of gga-miR-99a significantly depressed SMARCA5 expression, whereas a gga-miR-99a inhibitor enhanced the expression of SMARCA5. Inversely, SMARCA5 was significantly up-regulated and gga-miR-99a was obviously down-regulated in MG-HS-infected chicken embryonic lungs and DF-1 cells. At 72h post-transfection, the over-expression of gga-miR-99a significantly repressed the proliferation of DF-1 cells by inhibiting the transition from the G1 phase to the S and G2 phases. This study reveals that gga-miR-99a plays a key role in MG infection through the regulation of SMARCA5 expression and provides new insights regarding the mechanisms of MG pathogenesis.


Frontiers in Cellular and Infection Microbiology | 2016

Chicken gga-miR-19a Targets ZMYND11 and Plays an Important Role in Host Defense against Mycoplasma gallisepticum (HS Strain) Infection

Qingchang Hu; Yabo Zhao; Zaiwei Wang; Yue Hou; Dingren Bi; Jianjun Sun; Xiuli Peng

Mycoplasma gallisepticum (MG), one of the most pathogenic Mycoplasmas, can cause chronic respiratory disease (CRD) in chickens. It has been suggested that micro-ribonucleic acids (miRNAs) are involved in microbial pathogenesis. However, little is known about the roles of miRNAs in MG infection. Previously, we found by deep sequencing that gga-miR-19a was significantly up-regulated in the lungs of MG-infected chicken embryos. In this work, we confirmed that gga-miR-19a was up-regulated in both MG-infected chicken embryonic lungs and MG-infected DF-1 (chicken embryo fibroblast) cells. At 72 h post-transfection, we found that the over-expression of gga-miR-19a significantly enhanced the proliferation of MG-infected DF-1 cells by promoting the transition from the G1 phase to the S and G2 phases, while a gga-miR-19a inhibitor repressed the proliferation of MG-infected DF-1 cells by arresting the cell cycle in the G1 phase. Moreover, we found that gga-miR-19a regulated the expression of the host zinc-finger protein, MYND-type containing 11 (ZMYND11), through binding to its 3′ untranslated region (3′-UTR). DAVID analysis revealed that ZMYND11 could negatively regulate the NF-kappaB (NF-κB) signaling pathway in chickens (Gallus gallus). Upon MG infection, gga-miR-19a, NF-κB, MyD88, and TNF-α were all up-regulated, whereas ZMYND11 was down-regulated. The over-expression of gga-miR-19a in the DF-1 cells did not affect the above gene expression patterns, and gga-miR-19a inhibitor repressed the expression of NF-κB, MyD88, and TNF-α, but enhanced the expression of ZMYND11. In conclusion, gga-miR-19a might suppress the expression of ZMYND11 in MG-infected chicken embryonic lungs and DF-1 cells, activate the NF-κB signaling pathway, and promote pro-inflammatory cytokines expression, the cell cycle progression and cell proliferation to defend against MG infection.


International Journal of Molecular Sciences | 2018

gga-miR-451 Negatively Regulates Mycoplasma gallisepticum (HS Strain)-Induced Inflammatory Cytokine Production via Targeting YWHAZ

Yabo Zhao; Kang Zhang; Mengyun Zou; Yingfei Sun; Xiuli Peng

Mycoplasma gallisepticum (MG) is the most economically significant mycoplasma pathogen of poultry that causes chronic respiratory disease (CRD) in chickens. Although miRNAs have been identified as a major regulator effect on inflammatory response, it is largely unclear how they regulate MG-induced inflammation. The aim of this study was to investigate the functional roles of gga-miR-451 and identify downstream targets regulated by gga-miR-451 in MG infection of chicken. We found that the expression of gga-miR-451 was significantly up-regulated during MG infection of chicken embryo fibroblast cells (DF-1) and chicken embryonic lungs. Overexpression of gga-miR-451 decreased the MG-induced inflammatory cytokine production, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), whereas inhibition of gga-miR-451 had the opposite effect. Gene expression data combined with luciferase reporter assays demonstrated that tyrosine3-monooxygenase/tryptophan5-monooxygenase activation protein zeta (YWHAZ) was identified as a direct target of gga-miR-451 in the context of MG infection. Furthermore, upregulation of gga-miR-451 significantly inhibited the MG-infected DF-1 cells proliferation, induced cell-cycle arrest, and promoted apoptosis. Collectively, our results demonstrate that gga-miR-451 negatively regulates the MG-induced production of inflammatory cytokines via targeting YWHAZ, inhibits the cell cycle progression and cell proliferation, and promotes cell apoptosis. This study provides a better understanding of the molecular mechanisms of MG infection.

Collaboration


Dive into the Xiuli Peng's collaboration.

Top Co-Authors

Avatar

Jianjun Sun

University of Texas at El Paso

View shared research outputs
Top Co-Authors

Avatar

Yabo Zhao

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Kang Zhang

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Yue Hou

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Dingren Bi

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Zaiwei Wang

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Chengcheng Zhao

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Jiao Chen

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Wei Tian

Huazhong Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Dan Li

Huazhong Agricultural University

View shared research outputs
Researchain Logo
Decentralizing Knowledge