Network


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

Hotspot


Dive into the research topics where Ke Tao is active.

Publication


Featured researches published by Ke Tao.


Scientific Reports | 2016

Melatonin prevents acute kidney injury in severely burned rats via the activation of SIRT1

Xiaozhi Bai; Ting He; Jianxin Gao; Yang Liu; Jiaqi Liu; Shichao Han; Yan Li; Jihong Shi; Juntao Han; Ke Tao; Song-Tao Xie; Hongtao Wang; Dahai Hu

Acute kidney injury (AKI) is a common complication after severe burns. Melatonin has been reported to protect against multiple organ injuries by increasing the expression of SIRT1, a silent information regulator that regulates stress responses, inflammation, cellular senescence and apoptosis. This study aimed to investigate the protective effects of melatonin on renal tissues of burned rats and the role of SIRT1 involving the effects. Rat severely burned model was established, with or without the administration of melatonin and SIRT1 inhibitor. The renal function and histological manifestations were determined to evaluate the severity of kidney injury. The levels of acetylated-p53 (Ac-p53), acetylated-p65 (Ac-p65), NF-κB, acetylated-forkhead box O1 (Ac-FoxO1), Bcl-2 and Bax were analyzed to study the underlying mechanisms. Our results suggested that severe burns could induce acute kidney injury, which could be partially reversed by melatonin. Melatonin attenuated oxidative stress, inflammation and apoptosis accompanied by the increased expression of SIRT1. The protective effects of melatonin were abrogated by the inhibition of SIRT1. In conclusion, we demonstrate that melatonin improves severe burn-induced AKI via the activation of SIRT1 signaling.


Burns | 2013

Inhibition of Na+/H+ exchanger 1 by cariporide reduces burn-induced intestinal barrier breakdown.

Xuekang Yang; Ji Chen; Hua Bai; Ke Tao; Qin Zhou; Hongyi Hou; Dahai Hu

Severe burns initiate an inflammatory cascade within the gut, which leads to intestinal mucosal injury. Although Na(+)/H(+) exchanger 1 (NHE1) is recognised as a pivotal player in several inflammatory processes, its role in burn-induced intestinal injury is relatively unknown. We hypothesised that NHE1 might be involved in the increased intestinal permeability and barrier breakdown after severe burns. Thus, we here investigate whether the inhibition of NHE1 has a protective effect on burn-induced intestinal injury. Mice were subjected to a 30% total body surface area (TBSA) full-thickness steam burn. Cariporide was used to assess the function of NHE1 in mice with burn-induced intestinal injury by fluorescence spectrophotometry, Western blotting and enzyme linked immunosorbent assay (ELISA). We found that severe burn increased intestinal permeability, associated with the up-regulation of NHE1 and raised inflammatory cytokine levels. Mice treated with the NHE1 inhibitor cariporide had significantly attenuated burn-induced intestinal permeability and a reduced inflammatory response. NHE1 inhibition also reduced nuclear factor-κB (NF-κB) activation and attenuated p38 mitogen-activated protein kinase (MAPK) phosphorylation. Our study suggests that NHE1 plays an important role in burn-induced intestinal permeability through the regulation of the inflammatory response. Inhibition of NHE1 may be adopted as a potential therapeutic strategy for attenuating intestinal barrier breakdown.


American Journal of Physiology-cell Physiology | 2018

Acute down-regulation of miR-199a attenuates sepsis-induced acute lung injury by targeting SIRT1

Yang Liu; Hao Guan; Julei Zhang; Zhao Zheng; Hongtao Wang; Ke Tao; Shichao Han; Linlin Su; Dahai Hu

MicroRNA-199a (miR-199a) is a novel gene regulator with an important role in inflammation and lung injury. However, its role in the pathogenesis of sepsis-induced acute respiratory distress syndrome (ARDS) is currently unknown. Our study explored the role of miR-199a in sepsis-induced ARDS and its mechanism of action. First, we found that LPS could upregulate miR-199a in alveolar macrophages. Downregulation of miR-199a inhibited the upregulation of inflammatory cytokines in alveolar macrophages and induced the remission of histopathologic changes, the reduction of proinflammatory cytokines, and the upregulation of apoptosis protein expression in an ARDS lung, showing a protective role for miR-199a. We further identified sirtuin 1 (SIRT1) as a direct target of miR-199a in alveolar macrophages, and the expression of SIRT1 was negatively correlated with the level of miR-199a. The protective role of miR-199a downregulation in LPS-stimulated alveolar macrophages and sepsis-induced ARDS could be attenuated by SIRT1 inhibitor. Taken together, these results indicate that downregulation of miR-199a might protect lung tissue against sepsis-induced ARDS by upregulation of SIRT1 through the suppression of excessive inflammatory responses and the inhibition of cellular apoptosis in lung tissue, suggesting its potential therapeutic effects on sepsis-induced ARDS.


International Journal of Molecular Medicine | 2017

PKCζ as a promising therapeutic target for TNFα-induced inflammatory disorders in chronic cutaneous wounds

Jian Zhang; Xuekang Yang; Hongtao Wang; Bin Zhao; Xue Wu; Linlin Su; Song-Tao Xie; Yunchuan Wang; Jun Li; Jiaqi Liu; Mengdong Liu; Fu Han; Ting He; Wei Zhang; Ke Tao; Dahai Hu

Protein kinase Cζ (PKCζ) is a member of the atypical protein kinase C family. Its roles in macrophages or skin-resident keratinocytes have not been fully evaluated. In this study, we provide evidence that PKCζ mediates lipopolysaccharide (LPS)-induced tumor necrosis factor α (TNFα) gene expression in the mouse macrophage cell line, RAW264.7. TNFα has been proven to be one of the main culprits of chronic wounds and impaired acute wounds, which are characterized by excessive inflammation, enhanced proteolysis and reduced matrix deposition. Among the multiple effects of TNFα on keratinocytes, the induction of chemokines which are indispensable factors involved in the massive infiltration of various inflammatory cells into skin lesions serves as a crucial mechanism. In the present study, we found that PKCζ inhibitor or its specific siRNA inhibited the TNFα-induced upregulation in the levels of the chemokines, interleukin (IL)-8, monocyte chemotactic protein-1 (MCP-1) and intercellular cell adhesion molecule-1 (ICAM-1) in HaCaT keratinocytes. Moreover, under a disrupted inflammatory environment, activated keratinocytes can synthesize large amounts of matrix metalloproteinases (MMP), which has a negative effect on tissue remodeling. We discovered that TNFα promoted the expression of MMP9 in a PKCζ-dependent manner. Further experiments revealed that nuclear factor-κB (NF-κB) was a key downstream molecule of PKCζ. In addition, as shown in vitro, PKCζ was not involved in the TNFα-induced decrease in HaCaT cell migration and proliferation. In vivo experiments demonstrated that TNFα-induced wound closure impairment and inflammatory disorders were significantly attenuated in the PKCζ inhibitor group. On the whole, our findings suggest that PKCζ is a crucial regulator in LPS- or TNFα-induced inflammatory responses in RAW264.7 cells and HaCaT keratinocytes, and that PKCζ/NF-κB signaling may be a potential target for interventional therapy for TNFα-induced skin inflammatory injury.


Cell Cycle | 2017

Efficient generation of functional Schwann cells from adipose-derived stem cells in defined conditions.

Song-Tao Xie; Fan Lu; Juntao Han; Ke Tao; Hongtao Wang; Alfred Simental; Dahai Hu; Hao Yang

ABSTRACT Schwann cells (SCs) are hitherto regarded as the most promising candidates for viable cell-based therapy to peripheral nervous system (PNS) injuries or degenerative diseases. However, the extreme drawbacks of transplanting autologous SCs for clinical applications still represent a significant bottleneck in neural regenerative medicine, mainly owing to the need of sacrificing a functional nerve to generate autologous SCs and the nature of slow expansion of the SCs. Thus, it is of great importance to establish an alternative cell system for the generation of sufficient SCs. Here, we demonstrated that adipose-derived stem cells (ADSCs) of rat robustly give rise to morphological, phenotypic and functional SCs using an optimized protocol. After undergoing a 3-week in vitro differentiation, almost all of treated ADSCs exhibited spindle shaped morphology similar to genuine SCs and expressed SC markers GFAP and S100. Most importantly, apart from acquisition of SC antigenic and biochemical features, the ADSC-derived SCs were functionally identical to native SCs as they possess a potential ability to form myelin, and secret nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) and glia-derived neurotrophic factor (GDNF). The current study may provide an ideal strategy for harvesting sufficient SCs for cell-based treatment of various peripheral nerve injuries or disorders.


Experimental Cell Research | 2018

Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway

Wei Zhang; Xiaozhi Bai; Bin Zhao; Yan Li; Yijie Zhang; Zhenzhen Li; Xujie Wang; Liang Luo; Fu Han; Julei Zhang; Shichao Han; Weixia Cai; Linlin Su; Ke Tao; Jihong Shi; Dahai Hu

Introduction: Adipose tissue‐derived stem cells (ADSCs) have been shown to enhance wound healing via their paracrine function. Exosomes, as one of the most important paracrine factors, play an essential role in this process. However, the concrete mechanisms that underlie this effect are poorly understood. In this study, we aim to explore the potential roles and molecular mechanisms of exosomes derived from ADSCs in cutaneous wound healing. Methods: Normal human skin fibroblasts and ADSCs were isolated from patient skin and adipose tissues. ADSCs were characterized by using flow cytometric analysis and adipogenic and osteogenic differentiation assays. Exosomes were purified from human ADSCs by differential ultracentrifugation and identified by electron microscopy, nanoparticle tracking, fluorescence confocal microscopy and western blotting. Fibroblasts were treated with different concentrations of exosomes, and the synthesis of collagen was analyzed by western blotting; the levels of growth factors were analyzed by real‐time quantitative PCR (RT‐PCR) and ELISA; and the proliferation and migration abilities of fibroblasts were analyzed by real‐time cell analysis, CCK‐8 assays and scratch assays. A mouse model with a full‐thickness incision wound was used to evaluate the effect of ADSC‐derived exosomes on wound healing. The level of p‐Akt/Akt was analyzed by western blotting. Ly294002, a phosphatidylinositol 3‐kinases (PI3K) inhibitor, was used to identify the underlying mechanisms by which ADSC‐derived exosomes promote wound healing. Results: ADSC‐derived exosomes were taken up by the fibroblasts, which showed significant, dose‐dependent increases in cell proliferation and migration compared to the behavior of cells without exosome treatment. More importantly, both the mRNA and protein levels of type I collagen (Col 1), type III collagen (Col 3), MMP1, bFGF, and TGF‐&bgr;1 were increased in fibroblasts after stimulation with exosomes. Furthermore, exosomes significantly accelerated wound healing in vivo and increased the level of p‐Akt/Akt in vitro. However, Ly294002 alleviated these exosome‐induced changes, suggesting that exosomes from ADSCs could promote and optimize collagen deposition in vitro and in vivo and further promote wound healing via the PI3K/Akt signaling pathway. Conclusions: This study demonstrates that ADSC‐derived exosomes can promote fibroblast proliferation and migration and optimize collagen deposition via the PI3K/Akt signaling pathway to further accelerate wound healing. Our results suggest that ADSCs likely facilitate wound healing via the release of exosomes, and the PI3K/Akt pathway may play a role in this process. Our data also suggest that the clinical application of ADSC‐derived exosomes may shed new light on the use of cell‐free therapy to accelerate full‐thickness skin wound healing and attenuate scar formation. Graphical abstract Schematic diagram shows how the wound healing effect of ADSC‐Exos is mediated by the activation of the PI3K/Akt signaling pathways. Figure. No Caption available. HighlightsADSC‐Exos are internalized into HDFs and regulate their biological behaviors and functions.ADSC‐Exos play an important role in accelerating wound healing via activating PI3K/Akt signaling pathway.ADSC‐Exos may serve as a cell‐free therapy for the potential clinical treatment of wound healing.


Journal of Microbiological Methods | 2016

Expression and purification of rhIL-10-RGD from Escherichia coli as a potential wound healing agent.

Fangfang Yang; Yi Wan; Jiaqi Liu; Xuekang Yang; Hongtao Wang; Ke Tao; Juntao Han; Jihong Shi; Dahai Hu

Various protocols for recombinant Interleukin-10 (IL-10) purification in wound healing have been reported previously. However, the therapeutic effect was not obvious. Thus, it is of great importance to find new and effective approaches for therapy. In this study, we propose that IL-10 and Arginine-Glycine-Aspartic (RGD) peptide would be a valuable therapeutic for wound healing. To explore a high-efficiency and cost-effective approach for the production of IL-10 and RGD peptide with bioactivity, a synthetic gene was cloned into a recombinant pTWIN1 vector. As a consequence, rhIL-10-RGD and the pH-induced self-cleavable Ssp DnaB mini-intein as a fusion protein was highly expressed by IPTG induction in Escherichia coli Rosetta without extra residues in a bioreactor. After Ni affinity chromatographic purification, rhIL-10-RGD was released by the Ssp DnaB intein-mediated self-cleavage that is triggered by pH shift. SDS-PAGE and silver staining showed a major band with an estimated molecular mass of 19.3kDa. Cell proliferation assay confirmed its potent proliferation activity on MC/9 murine mast cells. In conclusion, we report a novel strategy to produce rhIL-10-RGD mediated by the pH-induced self-cleavable Ssp DnaB mini-intein, and show that rhIL-10-RGD could play an effective role in wound healing of BALB/c mice.


Immunology Letters | 2016

The amelioration of composite tissue allograft rejection by TIM-3-modified dendritic cell: Regulation of the balance of regulatory and effector T cells

Yaojun Wang; Zhao Zheng; Xiongxiang Zhu; Juntao Han; Maolong Dong; Ke Tao; Hongtao Wang; Yunchuan Wang; Dahai Hu

T cell-dependent immune responses play a central role in allograft rejection. Exploring ways to disarm alloreactive T cells represents a potential strategy to promote long-term allograft acceptance and survival. T cell Ig domain and mucin domain 3 (TIM-3) has previously been demonstrated as a central regulator of T helper 1 (Th1) responses and immune tolerance. Hence, TIM-3 may be an important molecule for decreasing immunological rejection during composite tissue allotransplantation (CTA). In this study, BALB/c and C57BL/6 mice were chosen as the experimental animals. The effects of TIM-3 on allograft rejection were explored using TIM-3-modified mature dendritic cells (TIM-3 mDCs). A laser speckle blood flow (LSBF) imager was used to evaluate blood distribution of the BALB/c mice. ELISA, MTT, ELISPOT assays and flow cytometry analysis were carried out for further researches. We found that TIM-3 could obviously prolong the survival time of the transplanted limbs. And TIM-3 could mitigate the immune response and thus enhance immune tolerance after CTA. Also, TIM-3 can induce lymphocyte hyporesponsiveness, including facilitating lymphocyte apoptosis, decreasing lymphocyte proliferation, and influencing the secretion of inflammatory cytokines by CD4(+) T cells. Furthermore, TIM-3 overexpression could induce CD4(+) T cells to differentiate into regulatory T cells (Tregs), which recalibrate the effector and regulatory arms of the alloimmune response. In summary, we concluded that TIM-3 can mitigate allograft rejection and thus enhance immune tolerance by inducing lymphocyte hyporesponsiveness and increasing the number of Tregs of the alloimmune response. TIM-3 may be a potential therapeutic molecule for allograft rejection in CTA.


Laboratory Investigation | 2018

Wild-type p53-modulated autophagy and autophagic fibroblast apoptosis inhibit hypertrophic scar formation

Jihong Shi; Houan Xiao; Jun Li; Julei Zhang; Yan Li; Jian Zhang; Xujie Wang; Xiaozhi Bai; Ke Tao; Dahai Hu; Hao Guan

Hypertrophic scarring is a serious fibrotic skin disease, and the abnormal activation of hypertrophic scar fibroblasts (HSFs) intensifies its pathogenesis. Our previous studies have demonstrated that the dysregulation of autophagy in HSFs is associated with fibrosis. However, knowledge regarding the regulation of HS fibrosis by p53-modulated autophagy is limited. Here, we investigated the effect of p53-modulated autophagy on HS fibrosis. The overexpression of wtp53 (Adp53) promoted autophagic capacity and inhibited collagen and α-SMA expression in HSFs. In contrast, LC3 (AdLC3) overexpression did not suppress Col 1, Col 3, or α-SMA expression, but LC3 (shLC3) knockdown downregulated collagen expression. Adp53-modulated autophagy altered Bcl-2 and Bcl-xL expression, but AdLC3 affected only Bcl-xL expression. Silencing Bcl-xL suppressed collagen expression, but autophagy was also inhibited. Flow cytometry showed that the silencing of Bcl-2 (sibcl-2), Bcl-xL (sibcl-xL), and Adp53 significantly increased apoptosis in the HSFs. Therefore, wtp53 inhibited fibrosis in the HSFs by modulating autophagic HSF apoptosis; moreover, the inhibition of autophagy by sibcl-xL had antifibrotic effects. In addition, treatment with Adp53, AdLC3, shLC3, sibcl-2, and sibcl-xL reduced scar formation in a rabbit ear scar model. These data confirm that wtp53-modulated autophagy and autophagic HSF apoptosis can serve as potential molecular targets for HS therapy.The dysregulation of dermal fibroblast proliferation and apoptosis is thought to be related to hypertrophic scar formation. In this study, the authors show that the balance of autophagy in HSF is critical for fibrosis formation, and both downregulation and excessive upregulation of autophagy inhibits collagen expression via wild type p53 and the pro-survival protein Bcl-xL.


Journal of Surgical Research | 2018

Free vascularized fascia flap combined with skin grafting for deep toe ulcer in diabetic patients

Xuekang Yang; Zhuoqun Fang; Mengdong Liu; Yue Zhang; Qiaohua Chen; Ke Tao; Juntao Han; Dahai Hu

BACKGROUNDnThis study introduces a technique for the reconstruction of deep toe defects in diabetic patients using a method that combines free vascularized fascia flap with skin grafting.nnnMETHODSnIn this retrospective study, conducted between March 2010 and February 2016, 15 diabetic patients with deep toe ulcer received surgeries that combined free vascularized fascia flap with skin grafting, including nine anterolateral thigh fascia lata flaps and six superficial temporal fascia flaps. Their medical records were systematically reviewed from electronic databases. The donor artery was anastomosed to the dorsalis pedis artery in an end-to-side manner, and the vein was anastomosed to the accompanying vein in an end-to-end manner.nnnRESULTSnThirteen fascia flaps completely survived without any rejection. Partially necrosed grafted skins, which were found in two cases, were healed after routine dressing changes. Patients achieved an esthetic outcome and acceptable functions without further revisions. Two patients suffered from ischemic necrosis of the fascia flap and eventually underwent amputation.nnnCONCLUSIONSnThe present study demonstrated that vascularized fascia flap combined with skin grafting has great advantages for correcting deep toe ulcer in diabetic patients characterized by the esthetic outcome, abundant vascularity, surgical simplicity, and good deformability.

Collaboration


Dive into the Ke Tao's collaboration.

Top Co-Authors

Avatar

Dahai Hu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongtao Wang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xuekang Yang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Juntao Han

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiaqi Liu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jihong Shi

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jun Li

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Song-Tao Xie

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaozhi Bai

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Julei Zhang

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge