Network


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

Hotspot


Dive into the research topics where Jihong Shi is active.

Publication


Featured researches published by Jihong Shi.


Stem Cell Research & Therapy | 2016

Adipose tissue-derived stem cells suppress hypertrophic scar fibrosis via the p38/MAPK signaling pathway

Yan Li; Wei Zhang; Jianxin Gao; Jiaqi Liu; Hongtao Wang; Jun Li; Xuekang Yang; Ting He; Hao Guan; Zhao Zheng; Shichao Han; Maolong Dong; Juntao Han; Jihong Shi; Dahai Hu

BackgroundHypertrophic scars (HS) generally occur after injury to the deep layers of the dermis, resulting in functional deficiency for patients. Growing evidence has been identified that the supernatant of adipose tissue-derived stem cells (ADSCs) significantly ameliorates fibrosis of different tissues, but limited attention has been paid to its efficacy on attenuating skin fibrosis. In this study, we explored the effect and possible mechanism of ADSC-conditioned medium (ADSC-CM) on HS.MethodReal-time quantitative polymerase chain reaction (qRT-PCR) and Western blotting were used to detect the expression of collagen I (Col1), collagen III (Col3), and α-smooth muscle actin (α-SMA) after fibroblasts and cultured HS tissues were stimulated with ADSC-CM and p38 inhibitor/activator. Immunofluorescence staining was performed to test the expression of α-SMA. Masson’s trichrome staining, hematoxylin and eosin (H&E) staining, and immunohistochemistry staining were carried out to assess the histological and pathological change of collagen in the BALB/c mouse excisional model. All data were analyzed by using SPSS17.0 software. Statistical analysis was performed by Student’s t tests.ResultsThe in vitro and ex vivo study revealed ADSC-CM decreased the expression of Col1, Col3, and α-SMA. Together, thinner and orderly arranged collagen was manifested in HS tissues cultured with ADSC-CM. Dramatically, the assessed morphology showed an accelerated healing rate, less collagen deposition, and col1- and col3-positive cells in the ADSC-CM treated group. Importantly, the protein level of p-p38 was downregulated in a concentration-dependent manner in HS-derived fibroblasts with ADSC-CM treatment, which further decreased the expression of p-p38 after the application of its inhibitor, SB203580. SB203580 led to an obvious decline in the expression of Col1, Col3, and α-SMA in fibroblasts and cultured HS tissues and presented more ordered arrangement and thinner collagen fibers in BALB/c mice. Lastly, anisomycin, an agonist of p38, upregulated the expression of fibrotic proteins and revealed more disordered structure and denser collagen fibers.ConclusionThis study demonstrated that ADSC-CM could decrease collagen deposition and scar formation in in vitro, ex vivo and in vivo experiments. The regulation of the p38/MAPK signaling pathway played an important role in the process. The application of ADSC-CM may provide a novel therapeutic strategy for HS treatment, and the anti-scarring effect can be achieved by inhibition of the p38/MAPK signaling pathway.


Cytotherapy | 2006

A mutated human tumor necrosis factor-alpha improves the therapeutic index in vitro and in vivo.

Zhen Yan; Ning Zhao; Zenglu Wang; B. Li; Chunjie Bao; Jihong Shi; Wei Han; Yingqi Zhang

BACKGROUND Tumor necrosis factor-alpha (TNF-alpha) is a multifunctional cytokine that has cytotoxic, cytostatic and immunomodulatory effects on malignant tumors. However, clinical trials have revealed high systemic toxicity and this has hampered its utilization as an anti-cancer agent. In this study, a human TNF-alpha mutant was created and tested for its anti-tumor effects. METHODS The TNF mutant (recombinant mutated human TNF; rmhTNF) was prepared by protein engineering in which amino acids Pro, Ser and Asp at positions 8, 9 and 10 of TNF-alpha were substituted by Arg, Lys and Arg, and C terminal Leu157 was substituted by Phe, along with deletion of the first seven N-terminal amino acids. Prokaryotic expression recombinant vector pBV-mhTNF containing the PLPR promotor was constructed and transformed into E. coli DH5alpha. The rmhTNF was expressed in a partially soluble form in DH5alpha, purified from the supernatant of cell lysate by ammonia sulfate precipitation and two sequential chromatographic steps. RESULTS The purified rmhTNF was >95% pure by SDS-PAGE stained with silver and high-pressure size exclusion chromatography (SEC-HPLC). Its yield was about 1.22 mg/g wet cell paste. The mutant rmhTNF exhibited an approximately 50-fold increase in cytotoxicity relative to the wild-type rhTNF on the mouse fibroblast cell line L929 in a standard cytotoxicity test, and at least and at least 50 times higher LD50 as wild type rhTNF in mice. In vivo biological activity studies carried out on tumor cell transplanted mice and nude mice also showed a more effective cytotoxicity of rmhTNF than rhTNF. DISCUSSION These results suggest that rmhTNF has potential for developing an effective anti-tumor reagent for some tumors.


International Journal of Molecular Medicine | 2016

Src promotes cutaneous wound healing by regulating MMP-2 through the ERK pathway.

Xue Wu; Longlong Yang; Zhao Zheng; Zhenzhen Li; Jihong Shi; Yan Li; Shichao Han; Jianxin Gao; Chaowu Tang; Linlin Su; Dahai Hu

Wound healing is a highly orchestrated, multistep process, and delayed wound healing is a significant symptomatic clinical problem. Keratinocyte migration and re-epithelialization play the most important roles in wound healing, as they determine the rate of wound healing. In our previous study, we found that Src, one of the oldest proto-oncogenes encoding a membrane-associated, non-receptor protein tyrosine kinase, promotes keratinocyte migration. We therefore hypothesized that Src promotes wound healing through enhanced keratinocyte migration. In order to test this hypothesis, vectors for overexpressing Src and small interfering RNAs (siRNAs) for silencing of Src were used in the present study. We found that the overexpression of Src accelerated keratinocyte migration in vitro and promoted wound healing in vivo without exerting a marked effect on cell proliferation. The extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling pathways play important roles in Src-accelerated keratinocyte migration. Further experiments demonstrated that Src induced the protein expression of matrix metallopro-teinase-2 (MMP-2) and decreased the protein expression of E-cadherin. We suggest that ERK signaling is involved in the Src-mediated regulation of MMP-2 expression. The present study provided evidence that Src promotes keratinocyte migration and cutaneous wound healing, in which the regulation of MMP-2 through the ERK pathway plays an important role, and thus we also demonstrated a potential therapeutic role for Src in cutaneous wound healing.


British Journal of Pharmacology | 2016

Identification of sirtuin 1 as a promising therapeutic target for hypertrophic scars

Xiaozhi Bai; Jiaqi Liu; Longlong Yang; Lei Fan; Ting He; Linlin Su; Jihong Shi; Chaowu Tang; Zhao Zheng; Dahai Hu

Sirtuin1 (SIRT1), the founding member of mammalian class III histone deacetylases, is reported to be a drug target involved in fibrotic diseases. However, whether it is an effective drug target in hypertrophic scar treatment is still not known.


Journal of Molecular Histology | 2017

MicroRNA-192 regulates hypertrophic scar fibrosis by targeting SIP1

Yan Li; Julei Zhang; Wei Zhang; Yang Liu; Yue-Hua Li; Kejia Wang; Yijie Zhang; Chen Yang; Xiaoqiang Li; Jihong Shi; Linlin Su; Dahai Hu

Hypertrophic scar (HS) is a fibro-proliferative disorder which is characterized by excessive deposition of collagen and accumulative activity of myofibroblasts. Increasing evidences have demonstrated miRNAs play a pivotal role in the pathogenesis of HS. MiR-192 is closely associated with renal fibrosis, but its effect on HS formation and skin fibrosis remains unknown. In the study, we presented that miR-192 was up-regulated in HS and HS derived fibroblasts (HSFs) compared to normal skin (NS) and NS derived fibroblasts (NSFs), accompanied by the reduction of smad interacting protein 1 (SIP1) expression and the increase of Col1, Col3 and α-SMA levels. Furthermore, we confirmed SIP1 was a direct target of miR-192 by using luciferase reporter assays. Meanwhile, the overexpression of miR-192 increased the levels of Col1, Col3 and α-SMA. The synthesis of collagen and more positive α-SMA staining were also observed in bleomycin-induced dermal fibrosis model of BALB/c mice treated with subcutaneous miR-192 mimics injection, whereas the inhibition of miR-192 decreased the expression of Col1, Col3 and α-SMA. Moreover, SIP1 siRNA could enhance the levels of Col1, Col3 and α-SMA, showing that the effect of knockdown SIP1 was similar to miR-192 mimics, and the phenomenon manifested miR-192 regulated HS fibrosis by targeting SIP1. Together, our results indicated that miR-192 was a critical factor of HS formation and facilitated skin fibrosis by targeting directly SIP1.


Applied Biochemistry and Biotechnology | 2006

Expression, refolding, and characterization of GFE peptide-fused human interferon-α2a in Escherichia coli

Zhen Yan; Li Lu; Jihong Shi; Chunjie Bao; Wei Han; Yongjie Wu; Yingqi Zhang

Interferon-α2a (IFN-α2a) has been used for the treatment of various viral infections and cancers for many years. However some untolerable side effects have limited its application in some aspects. To evaluate whether or not an oligopeptide containing GFE motif can home human IFN-α2a to specific tissues, a fusion gene was constructed by fusing the coding sequence of GFE peptide (CGFECVRQCPERC), which was screened from phage display peptide library, to the 3′ end of human IFN-α2a gene by recombinant DNA technique. Fusion protein rhIFN-α2a-GFE was expressed in Escherichia coli as inclusion bodies using a T7 RNA polymerase expression system, pET-22b, refolded through dialysis and purified to homogeneity to >95% of purity by affinity chromatography. Characterization by sodium dodecyl sulfate polyacrylamide gel electrophoresis and immunoblotting demonstrated the authenticity of the fusion protein. Purified rhIFN-α2a-GFE was found to be functionally active in terms of its antiviral activity for about 2.5×108 IU/mg in vitro. Yields of the purified fusion protein were about 200 mg/L of culture medium. Tissue distribution assay in mouse showed that at 30 min IFN-α2a could be enriched sevenfold higher in lung in the targeted IFN group of mice than in the standard IFN group of mice, and last for a long time. At 1 h, IFN-α2a in the targeted IFN group was still 4.02-fold higher than that in the standard group. This confirmed that GFE peptide has the ability to selectively deliver its fusion partner IFN-α2a to lungs. The results also showed that the IFN-α2a-GFE could be specifically enriched in kidney and liver. Its distribution in kidney was concordant with the finding of GFE receptor, MDP, in kidney. However, the IFN-α2a-GFE in liver may imply some significance in pharmacology and toxicology.


Biotechnology and Applied Biochemistry | 2003

Construction of a new tumour necrosis factor fusion‐protein expression vector for high‐level expression of heterologous genes in Escherichia coli

Wei Han; Yingqi Zhang; Zhen Yan; Jihong Shi

We report the construction and application of a new fusion‐protein expression plasmid (TNFHis) for Escherichia coli . The plasmid contains both PR and PL promoters and is optimized to allow a higher level of expression of mature coding sequences. It also contains a six‐histidine tag for convenient purification as well as thrombin and hydroxylamine recognition sites for cleaving heterologous protein. The potential use of this expression vector is demonstrated by comparing the expression levels of human tumour necrosis factor (TNF), interferon, interleukin 11, colony‐forming factor, osteoprotegrin and interleukin 2 in E. coli . Furthermore, all expressed TNF fusion proteins can be detected by anti‐TNFα antibody or by specific antibodies and purified by Ni2+‐nitrilotriacetate beads. The expressed TNF fusion proteins can be cleaved by hydroxylamine.


Frontiers in Immunology | 2018

Acetylation-Dependent Regulation of Notch Signaling in Macrophages by SIRT1 Affects Sepsis Development

Xiaozhi Bai; Ting He; Yang Liu; Julei Zhang; Xiaoqiang Li; Jihong Shi; Kejia Wang; Fu Han; Wei Zhang; Yijie Zhang; Weixia Cai; Dahai Hu

SIRT1 is reported to participate in macrophage differentiation and affect sepsis, and Notch signaling is widely reported to influence inflammation and macrophage activation. However, the specific mechanisms through which SIRT1 regulates sepsis and the relationship between SIRT1 and Notch signaling remain poorly elucidated. In this study, we found that SIRT1 levels were decreased in sepsis both in vitro and in vivo and that SIRT1 regulation of Notch signaling affected inflammation. In lipopolysaccharide (LPS)-induced sepsis, the levels of Notch signaling molecules, including Notch1, Notch2, Hes1, and intracellular domain of Notch (NICD), were increased. However, NICD could be deacetylated by SIRT1, and this led to the suppression of Notch signaling. Notably, in macrophages from myeloid-specific RBP-J−/− mice, in which Notch signaling is inhibited, pro-inflammatory cytokines were expressed at lower levels than in macrophages from wild-type littermates and in RBP-J−/− macrophages, and the NF-κB pathway was also inhibited. Accordingly, in the case of RBP-J−/− mice, LPS-induced inflammation and mortality were lower than in wild-type mice. Our results indicate that SIRT1 inhibits Notch signaling through NICD deacetylation and thus ultimately alleviates sepsis.


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.

Collaboration


Dive into the Jihong Shi's collaboration.

Top Co-Authors

Avatar

Dahai Hu

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
Top Co-Authors

Avatar

Yan Li

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Shichao Han

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Linlin Su

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Han

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yijie Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yingqi Zhang

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge