Network


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

Hotspot


Dive into the research topics where Chenggang Yi is active.

Publication


Featured researches published by Chenggang Yi.


The Lancet | 2008

Human facial allotransplantation: a 2-year follow-up study.

Shuzhong Guo; Yan Han; Xudong Zhang; Binglun Lu; Chenggang Yi; Hui Zhang; Xianjie Ma; Datai Wang; Li Yang; Xing Fan; Yunjing Liu; Kaihua Lu; Huiyuan Li

BACKGROUND Progress in composite tissue allotransplantation could provide a new treatment for patients with severe facial disfigurements. We did a partial facial allotransplantation in 2006, and report here the 2 year follow-up of the patient. METHODS The recipient, a 30-year-old man from China, had his face severely injured by a bear in October, 2004. Allograft composite tissue transplantation was done in April, 2006, after careful systemic preparation. The surgery included anastomosis of the right mandibular artery and anterior facial vein, whole repair of total nose, upper lip, parotid gland, front wall of the maxillary sinus, part of the infraorbital wall, and zygomatic bone. Facial nerve anastomosis was done during the surgery. Quadruple immunomodulatory therapy was used, containing tacrolimus, mycophenolate mofetil, corticosteroids, and humanised IL-2 receptor monoclonal antibody. Follow-up included T lymphocyte subgroups in peripheral blood, pathological and immunohistochemical examinations, functional progress, and psychological support. FINDINGS Composite tissue flap survived well. There were three acute rejection episodes at 3, 5, and 17 months after transplantation, but these were controlled by adjustment of the tacrolimus dose or the application of methylprednisolone pulse therapy. Hepatic and renal functions were normal, and there was no infection. The patient developed hyperglycaemia on day 3 after transplantation, which was controlled by medication. INTERPRETATION Facial transplantation could be successful in the short term, but the procedure was not without complications. However, promising results could mean that this procedure might be an option for long-term restoration of severe facial disfigurement.


Plastic and Reconstructive Surgery | 2008

Mesenchymal stem cells transduced by vascular endothelial growth factor gene for ischemic random skin flaps.

Yan Zheng; Chenggang Yi; Wei Xia; Tan Ding; Zhiqiang Zhou; Yan Han; Shuzhong Guo

Background: Vascular endothelial growth factor (VEGF) plays an important role in inducing angiogenesis. Mesenchymal stem cells may have the potential for differentiation into several types of cells, including vascular endothelial cells. In this study, the authors explored the feasibility of applying mesenchymal stem cells transduced by the VEGF gene to the treatment of ischemic random skin flaps. Methods: Mesenchymal stem cells were isolated from Sprague-Dawley rat bone marrow and cultured in vitro. Plasmid pcDNA3.1(-)/VEGF165 containing the VEGF gene was transduced into the mesenchymal stem cells by liposome. The mesenchymal stem cells were stained with chloromethyl-1-1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanineperchlorate before the transplantation. Thirty rats were randomized into three groups. Groups A, B, and C were injected with mesenchymal stem cells transduced with pcDNA3.1(-)/VEGF165 plasmid, mesenchymal stem cells, and medium only, respectively. On the fourth day after injection, random dorsal skin flaps measuring 9 × 2 cm were elevated. The survival, neovascularization, and blood flow recovery of the flaps were detected. Results: VEGF-transduced mesenchymal stem cells expressed VEGF highly in vitro and in vivo. Transplanted mesenchymal stem cells survived and incorporated into the capillary networks in the ischemic rat flaps. The viability measurements showed an increased percentage flap survival in group A (83.1 ± 2.6 percent) as compared with either group B (66.4 ± 6.1 percent) or group C (51.5 ± 7.5 percent)(p < 0.01). The capillary density and the blood perfusion of the flaps in the experimental group were significantly higher than those in the other two groups (p < 0.01). Conclusion: VEGF-transduced mesenchymal stem cells can increase ischemic flap neovascularization and augment the surviving areas.


Archives of Dermatological Research | 2011

Trichostatin A inhibits collagen synthesis and induces apoptosis in keloid fibroblasts

Jian-Sheng Diao; Wen-Sen Xia; Chenggang Yi; Ying-Mei Wang; Bing Li; Wei Xia; Bei Liu; Shuzhong Guo; Xu-De Sun

Keloid, a fibro-proliferative benign tumor of skin, is characterized by an enriched milieu of growth factors and an abundant accumulation of extracellular matrix (ECM). Transforming growth factor (TGF)-β1 is well known as the crucial fibrogenic cytokine promoting ECM production and tissue fibrosis in keloid forming. Epigenetic modifications have been shown to play a role in the pathogenesis of cancer as well as autoimmune and inflammatory disorders. Recent publication reports epigenetic modifications in keloid fibroblasts that include an altered pattern of DNA methylation and histone acetylation. Therefore, the field of epigenetics may provide a new therapeutic idea for keloid treatment strategies. Currently, there is some evidence from experimental studies that histone deacetylase (HDAC) inhibitor Trichostatin A (TSA) causes abrogation of TGF-β1 induced collagen synthesis in skin fibroblasts. Furthermore, TSA could suppress proliferation and induce apoptosis in a broad spectrum of tumor cells both in vitro and in vivo. These findings suggest that TSA could also cause abrogation of TGF-β1 induced collagen synthesis and induce apoptosis of proliferating keloid fibroblasts.


British Journal of Dermatology | 2009

Troglitazone suppresses transforming growth factor-β1-induced collagen type I expression in keloid fibroblasts

Guo-You Zhang; Chenggang Yi; Xia Li; Ma B; Zhijie Li; Xinglong Chen; Shuzhong Guo; Wei-Yang Gao

Background  Peroxisome proliferator‐activated receptor (PPAR)‐γ agonists are increasingly used in patients with diabetes and some studies have suggested a beneficial effect on organ fibrosis. However their effects on dermal fibrosis in keloids are unknown.


Archives of Dermatological Research | 2008

Inhibition of vascular endothelial growth factor expression in keloid fibroblasts by vector-mediated vascular endothelial growth factor shRNA : a therapeutic potential strategy for keloid

Guo-You Zhang; Chenggang Yi; Xuan Li; Yan Zheng; Zhan-Guo Niu; Wei Xia; Zhou Meng; Cheng-Yue Meng; S.Z. Guo

Vascular endothelial growth factor (VEGF) plays important roles in the regulation of angiogenesis and inflammation in both pathological and physiological wound repair. Several strategies have been developed for keloid therapy; however, a universally effective treatment has not been explored to date. In this study, three potential short interfering RNA (siRNA) sequences for the VEGF gene were cloned into expression plasmids and transfected into keloid fibroblasts. PGC-VEGF shRNA 2 (short hairpin RNA 2) plasmid significantly inhibited VEGF expression determined by real-time polymerase chain reaction (PCR), enzyme-linked immunosorbent assay (ELISA), and fibroblasts growth was also significantly by (methyl thiazolyl tetrazolium) MTT assay and apoptosis detection, whereas the control transfection showed no obviously effects. Plasminogen activator inhibitor-1 (PAI-1) expression in pGC-VEGF shRNA2 group was also obviously downregulated when compared to the pGC-VEGF shRNA negative control and mock group. These results suggest that modulation of VEGF production by vector-based RNAi in keloid fibroblasts may be a therapeutic potential strategy for keloid.


Archives of Dermatological Research | 2009

Activation of peroxisome proliferator-activated receptor-γ inhibits transforming growth factor-β1 induction of connective tissue growth factor and extracellular matrix in hypertrophic scar fibroblasts in vitro

Guo-You Zhang; Tao Cheng; Ming-Hua Zheng; Chenggang Yi; Hua Pan; Zhijie Li; Xinglong Chen; Qing Yu; Liang-Fu Jiang; Feiya Zhou; Xiao-Yang Li; Jingquan Yang; Tinggang Chu; Wei-Yang Gao

Peroxisome proliferator-activated receptor-γ (PPAR-γ) ligands have been recently reported to have beneficial effects on organ fibrosis. However, their effects on extracellular matrix (ECM) turnover in hypertrophic scar fibroblasts (HSFs), and the related molecular mechanisms are unknown. HSFs were cultured and exposed to different concentration PPAR-γ ligands in the presence of transforming growth factor-β1 (TGF-β1). In growth-arrested HSFs, a PPAR-γ natural ligand (15-deoxy-D12,14-prostaglandin J2, 15d-PGJ2) and a synthetic ligand (GW7845) dose-dependently attenuated TGFβ1-induced expression of Connective tissue growth factor (CTGF), collagens and fibronectin. Furthermore, the suppression of CTGF mRNA and protein expression are relieved by pretreatment with an antagonist of PPAR-γ (GW9662). Moreover, GW7845 and 15d-PGJ2 partially inhibited the expression and phosphorylation of the TGF-β1/Smad pathway. These results suggest that in TGFβ1-stimulated HSFs, PPAR-γ ligands caused an antiproliferative effect and reduced ECM production through mechanisms that included reducing CTGF expression, and a crosstalk between PPAR-γ and Smad may be involved in the inhibitory effects of PPAR-γ ligands.


Experimental Dermatology | 2009

Angiotensin II activates connective tissue growth factor and induces extracellular matrix changes involving Smad/activation and p38 mitogen-activated protein kinase signalling pathways in human dermal fibroblasts

Guo-You Zhang; Xuan Li; Chenggang Yi; Hua Pan; Guo-Dong He; Qing Yu; Liang-Fu Jiang; Wei-Hua Xu; Zhi-Jie Li; Jian Ding; Ding-Sheng Lin; Wei-Yang Gao

Abstract:  Angiotensin II (Ang II) stimulation has been shown to regulate proliferation of skin fibroblasts and the production of extracellular matrix, which are very important processes in skin wound healing and fibrosis; however, there is little knowledge about the mechanisms involved in this process. We investigated the molecular aspects of this system with regards to Ang II in human dermal fibroblasts (HDF) and its potential role in fibrosis. Fibroblasts derived from human skin were subjected to examine differential relative gene and protein expression after transfection with specific reporter expression vectors and Ang II in vitro. In growth‐arrested HDFs, Ang II treatment for 20 min caused acute activation of Smad2 phosphorylation, Smad overexpression and Smad‐dependent gene transcription. The angiotensin type 1 (AT1) antagonist losartan diminished Ang II‐induced Smad activation. The blockade of endogenous transforming growth factor‐β1 did modify the activation of Smad caused by Ang II. The p38 mitogen‐activated protein kinase (MAPK) inhibitor SB203580 diminished Ang II‐induced Smad2 phosphorylation. Transient transfection with Smad7, which interferes with receptor‐mediated activation of Smad2, diminished Ang II‐induced connective tissue growth factor promoter activation, gene and protein expression and fibronectin, type I procollagen and type III procollagen overexpression, showing that Smad activation is involved in Ang II‐induced dermal fibrosis. Our results show that Ang II activation of Smad2 occurs via the AT1 receptor, but not the AT2 receptor. Activation of Smad2 required p38 MAPK but not p42/p44 MAPK or the epidermal growth factor receptor.


Plastic and Reconstructive Surgery | 2012

Observations on the survival and neovascularization of fat grafts interchanged between C57BL/6-gfp and C57BL/6 mice.

Jianhui Zhao; Chenggang Yi; Long Li; Yan Zheng; Kangkang Wu; Lihua Liang; Wei Xia; Shuzhong Guo

Background: Autologous fat transplantation has become a prevalent option for soft-tissue augmentation throughout the body. However, there is still much controversy over whether the fat grafts have survived or have been replaced in the recipient sites and over how the vessels grow. Methods: After C57BL/6-gfp mice and C57BL/6 mice were paired randomly, the inguinal fat was excised and cut into pieces with scissors, and the adipose granules, approximately 0.2 ml (0.195 g), were transplanted subcutaneously with syringes to the dorsa of the paired mice. Samples were obtained at different time intervals: 3 days, 7 days, 2 weeks, 4 weeks, 2 months, 3 months, and 4 months after transplantation. Each fat sample was weighed to evaluate the graft volume. Histology, origin, and densities of neovascularization were examined by immunohistochemical staining. Results: At 4 months, there was no significant difference in either graft survival or histologic evaluation. Histologic evaluation manifested the normal physiologic process of inflammation, neovascularization, remodeling, and maturity at different time intervals. At the endpoint, the immunohistochemical staining of CD34 showed that the difference in capillary density of the fat graft—31.3 ± 3.9 capillaries/mm2 on the dorsa of the C57BL/6-gfp mice and 29.6 ± 3.2 capillaries/mm2 on the dorsa of the C57BL/6 mice—was not statistically significant. The &agr;-smooth muscle actin staining indicated that there were neovascularized vessels in both C57BL/6-gfp and C57BL/6 fat grafts. Conclusions: Fat grafts can survive and neovascularized vessels can grow from the recipient sites. Fat transplantation is feasible and will be applied more widely if fat graft survival is improved.


Journal of Plastic Reconstructive and Aesthetic Surgery | 2010

Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist inhibits transforming growth factor-beta1 and matrix production in human dermal fibroblasts

Guo-You Zhang; Tao Cheng; Ming-Hua Zheng; Chenggang Yi; Hua Pan; Zhijie Li; Xinglong Chen; Qing Yu; Liang-Fu Jiang; Feiya Zhou; Xiao-Yang Li; Jingquan Yang; Tinggang Chu; Wei-Yang Gao

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists are increasingly used in patients with diabetes, and some studies have suggested a beneficial effect on organ fibrosis, but their effects on dermal cell growth and extracellular matrix (ECM) turnover are unknown. To investigate the effect of the PPAR-gamma agonist troglitazone on cell growth and matrix production in human dermal fibroblasts (HDF), HDF were cultured and grown in a different concentration of troglitazone. PPAR-gamma expression and matrix production were measured in HDF in the presence of troglitazone. The mRNA expressions of TGF-beta1, collagen I (Col I) and fibronectin (FN) were determined by quantitative real-time reverse transcription polymerase chain reaction (RT-PCR). The protein of transforming growth factor-beta1 (TGF-beta1) was determined by enzyme-linked immunosorbent assay (ELISA) and proteins of Col I and FN were determined by Western blotting. The mRNA expression of TGF-beta1, Col I and FN were significantly decreased in HDF in 15-30 micromol l(-1) troglitazone compared to the control group with Dulbeccos modified Eagles medium (P<0.01). An obvious decrease of TGF-beta1 protein was found in troglitazone-treated groups as compared to the control group (P<0.05). Exposure of HDF to troglitazone reduced col I secretion (P<0.05), and fibronectin secretion (P<0.05). This study suggests that PPAR-gamma agonist will provide a novel approach with therapeutic potential in dermal fibrosis, such as hypertrophic scar, keloid and so on.


Medical Hypotheses | 2008

Possibilities and potential roles of estrogen in the pathogenesis of proliferation hemangiomas formation

Zhi-Yong Sun; Li Yang; Chenggang Yi; Huan Zhao; Da-Li Han; Tao Yang; Lu Wang; Chunlei Nie; Guo-You Zhang; Guo-Qian Yin; Gang Wang; Xiao-Pin Teng; Dong-Mei Fei; Jin Wang; Wen-Kai Zhou; Yang Li; Bin Liu; Yin Liu; Man-Jing Zhang; Shang-Min Wu; Xi Zhang; Hua Pan; Bo Xiao; Kefeng Zhao; Dan Liu; Shuzhong Guo

Hemangiomas, often categorized as angiogenic diseases, are the most common tumors of infancy, the life span of which is generally divided into proliferating phase, involuting phase, and involuted phase. Despite their high prevalence, the mechanism leading to proliferation hemangiomas formation is poorly understood and the best approach to their management remains controversial. None of the current therapeutic modalities is ideal, partly because the pathogenesis of hemangioma and the mechanism of its proliferation are far from clear. Many clues reveal that estrogen has an important role in developing the vascular system, experimental and clinical evidences accumulated in recent years also suggest the potential for estrogen to influence neovascularization. Based on those, we hypothesize that estrogen play a potential role in the development of hemangiomas, mainly by regulating some key angiogenic factors, including MMP-9, EPCs, VEGF, NO, etc. Accepting the hypothesis to be correct, a therapy that identify estrogen as a potential target for the design of new, more specific treatments can be used to prevent the proliferation hemangiomas formation. The hypothesis may lead a new direction in the study of mechanisms for proliferation hemangiomas formation, and further study of the precise mechanisms for estrogen-induced hemangiomas will produce effective antiestrogens and estrogen receptor antagonists as new medication for the very difficult problem.

Collaboration


Dive into the Chenggang Yi's collaboration.

Top Co-Authors

Avatar

Shuzhong Guo

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yingjun Su

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Xia

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Lihong Qiu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xianjie Ma

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Guo-You Zhang

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan Han

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan Zheng

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hui Zheng

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Kaihua Lu

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge