1
|
Zhao Z, Hu B, Zhang D, Cui X, Ding W, Wang T, Wang J, Guo Z, Zhou Y, Wang S, Guo J. Engineering cuttlefish melanin nanoparticles: A dual-action therapy for acute radiation syndrome and combined radiation-wound injuries via regulating Bcl-2 family proteins and caspases in the apoptotic process. CHEMICAL ENGINEERING JOURNAL 2025; 511:162108. [DOI: 10.1016/j.cej.2025.162108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
|
2
|
Zhang Y, Chen Y, Li K, Chen C, Hu Y, Li X. Ghrelin promotes chronic diabetic wound healing by regulating keratinocyte proliferation and migration through the ERK1/2 pathway. Peptides 2025; 184:171350. [PMID: 39824309 DOI: 10.1016/j.peptides.2025.171350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/05/2025] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
Delayed wound healing is a complication of diabetes mellitus and can lead to infection, sepsis, and amputation. Despite the currently available treatments, the global burden of diabetes-related wounds is growing; thus, more effective therapy for diabetic wounds is urgently needed. Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, is a 28-amino acid peptide hormone. Some reports have confirmed the therapeutic effects of ghrelin on diabetes mellitus and its complications. However, the effects and corresponding mechanisms of ghrelin on chronic diabetic wounds remain unknown. In this study, we explored the effect of ghrelin on diabetic wound healing and investigated the associated mechanisms. We showed that ghrelin accelerated wound healing in diabetic rats by promoting the proliferation and migration of keratinocytes. Re-epithelialization was accelerated in ghrelin-treated wounds, thicker and longer newly formed epidermis and more dividing keratinocytes were observed. We further confirmed that ghrelin regulated keratinocytes by activating the ERK1/2 pathway through its receptor growth hormone secretagogue receptor 1a (GHSR1a). Ghrelin also significantly reduced the levels of pro-inflammatory cytokines and increased the deposition of collagen in diabetic wounds. Our data provides preclinical evidence for the potential application of ghrelin as a compound to promote diabetic wound healing and clarifies the molecular mechanism.
Collapse
Affiliation(s)
- Yukang Zhang
- Department of Hand and Foot Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Yuan Chen
- Department of Central Research Lab, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Kailin Li
- Department of Central Research Lab, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Cong Chen
- Department of Hand and Foot Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Yong Hu
- Department of Hand and Foot Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Xian Li
- Department of Hand and Foot Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250033, China.
| |
Collapse
|
3
|
Li D, Lu Y, Xiao F, Cheng X, Hu C, Zhu X, Wang X, Duan H, Du L, Zhang Q. A recombinant plasmid encoding human hepatocyte growth factor promotes healing of combined radiation-trauma skin injury involved in regulating Nrf2 pathway in mice. JOURNAL OF RADIATION RESEARCH 2024; 65:279-290. [PMID: 38682896 PMCID: PMC11115442 DOI: 10.1093/jrr/rrae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/01/2024] [Indexed: 05/01/2024]
Abstract
Combined radiation-trauma skin injury represents a severe and intractable condition that urgently requires effective therapeutic interventions. In this context, hepatocyte growth factor (HGF), a multifunctional growth factor with regulating cell survival, angiogenesis, anti-inflammation and antioxidation, may be valuable for the treatment of combined radiation-trauma injury. This study investigated the protective effects of a recombinant plasmid encoding human HGF (pHGF) on irradiated human immortalized keratinocytes (HaCaT) cells in vitro, and its capability to promote the healing of combined radiation-trauma injuries in mice. The pHGF radioprotection on irradiated HaCaT cells in vitro was assessed by cell viability, the expression of Nrf2, Bcl-2 and Bax, as well as the secretion of inflammatory cytokines. In vivo therapeutic treatment, the irradiated mice with full-thickness skin wounds received pHGF local injection. The injuries were appraised based on relative wound area, pathology, immunohistochemical detection, terminal deoxynucleotidyl transferase dUTP nick end labelling assay and cytokine content. The transfection of pHGF increased the cell viability and Nrf2 expression in irradiated HaCaT cells. pHGF also significantly upregulated Bcl-2 expression, decreased the Bax/Bcl-2 ratio and inhibited the expression of interleukin-1β and tumor necrosis factor-α in irradiated cells. Local pHGF injection in vivo caused high HGF protein expression and noticeable accelerated healing of combined radiation-trauma injury. Moreover, pHGF administration upregulated Nrf2, vascular endothelial growth factor, Bcl-2 expression, downregulated Bax expression and mitigated inflammatory response. In conclusion, the protective effect of pHGF may be related to inhibiting apoptosis and inflammation involving by upregulating Nrf2. Local pHGF injection distinctly promoted the healing of combined radiation-trauma injury and demonstrates potential as a gene therapy intervention for combined radiation-trauma injury in clinic.
Collapse
Affiliation(s)
- Dujuan Li
- Department of Pharmacy & Pharmacology, University of South China, 28 Changsheng West Road, Zhengxiang District, Hengyang, Hunan 421001, China
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Yuxin Lu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Fengjun Xiao
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Xiaochen Cheng
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Chunsheng Hu
- Department of Pharmacology, College of Pharmacy & International Academy of Targeted Therapeutics and Innovation, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing University of Arts and Sciences, 319 Honghe avenue, Yongchuan District, Chongqing 402160, China
| | - Xuefeng Zhu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Xiaoying Wang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Haiying Duan
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Li Du
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Qinglin Zhang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| |
Collapse
|
4
|
Sharma AK, Kalonia A, Kumar R, Kirti, Shaw P, Yashvarddhan MH, Vibhuti A, Shukla SK. Alleviation of radiation combined skin injury in rat model by topical application of ascorbate formulation. Int J Radiat Biol 2024; 100:689-708. [PMID: 38306495 DOI: 10.1080/09553002.2024.2310016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
PURPOSE This research endeavor was undertaken to elucidate the impact of an innovative ascorbate formulation on the regeneration process of full-thickness excision wounds in a rat model exposed to whole-body gamma irradiation, replicating conditions akin to combat or radiation emergency scenarios. MATERIALS AND METHODS We established a comprehensive rat model by optimizing whole body γ-radiation doses (5-9 Gy) and full-thickness excision wound sizes (1-3 cm2) to mimic radiation combined injury (RCI). The developed RCI model was used to explore the healing potential of ascorbate formulation. The study includes various treatment groups (i.e., sham control, radiation alone, wound alone, radiation + wound, and radiation + wound + formulation). The ascorbate formulation was applied twice daily, with a 12-hour gap between each application, starting 1 hour after the initiation of the wound. The healing potential of the formulation in the RCI context was evaluated over 14 days through hematological, molecular, and histological parameters. RESULTS The combination of a 5 Gy radiation dose and a 1 cm2 wound was identified as the optimal setting to develop the RCI model for subsequent studies. The formulation was used topically immediately following RCI, and then twice daily until complete healing. Treatment with the ascorbate formulation yielded noteworthy outcomes and led to a substantial reduction (p < .05) in the wound area, accelerated epithelialization periods, and an increased wound contraction rate. The formulation's localized healing response improved organ weights, normalized blood parameters, and enhanced hematopoietic and immune systems. A gene expression study revealed the treatment up-regulated TGF-β and FGF, and down-regulated PDGF-α, TNF-α, IL-1β, IL-6, MIP-1α, and MCP-1 (p < .05). Histopathological assessments supported the formulation's effectiveness in restoring cellular architecture and promoting tissue regeneration. CONCLUSION Topical application of the ascorbate formulation in RCI resulted in a significant improvement in delayed wound healing, leading to accelerated wound closure by mitigating the expression of inflammatory responses.
Collapse
Affiliation(s)
- Ajay Kumar Sharma
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Aman Kalonia
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Rishav Kumar
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Kirti
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Priyanka Shaw
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - M H Yashvarddhan
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Arpana Vibhuti
- Department of Biotechnology, SRM University, Sonipat, Haryana, India
| | - Sandeep Kumar Shukla
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| |
Collapse
|
5
|
Shen J, Jiao W, Chen Z, Wang C, Song X, Ma L, Tang Z, Yan W, Xie H, Yuan B, Wang C, Dai J, Sun Y, Du L, Jin Y. Injectable multifunctional chitosan/dextran-based hydrogel accelerates wound healing in combined radiation and burn injury. Carbohydr Polym 2023; 316:121024. [PMID: 37321722 DOI: 10.1016/j.carbpol.2023.121024] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/17/2023]
Abstract
Clinical wound management of combined radiation and burn injury (CRBI) remains a huge challenge due to serious injuries induced by redundant reactive oxygen species (ROS), the accompanying hematopoietic, immunologic suppression and stem cell reduction. Herein, the injectable multifunctional Schiff base cross-linked with gallic acid modified chitosan (CSGA)/oxidized dextran (ODex) hydrogels were rationally designed to accelerate wound healing through elimination of ROS in CRBI. CSGA/ODex hydrogels, fabricated by mixing solutions of CSGA and Odex, displayed good self-healing ability, excellent injectability, strong antioxidant activity, and favorable biocompatibility. More importantly, CSGA/ODex hydrogels exhibited excellent antibacterial properties, which is facilitated for wound healing. Furthermore, CSGA/ODex hydrogels significantly suppressed the oxidative damage of L929 cells in an H2O2-induced ROS microenvironment. The recovery of mice with CRBI in mice demonstrated that CSGA/ODex hydrogels significantly reduced the hyperplasia of epithelial cells and the expression of proinflammatory cytokine, and accelerated wound healing which was superior to the treatment with commercial triethanolamine ointment. In conclusion, the CSGA/ODex hydrogels as a wound dressing could accelerate the wound healing and tissue regeneration of CRBI, which provides great potential in clinical treatment of CRBI.
Collapse
Affiliation(s)
- Jintao Shen
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wencheng Jiao
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Hebei University, Baoding 071002, China
| | - Ziyuan Chen
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chunqing Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xingshuang Song
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lei Ma
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ziyan Tang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrui Yan
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hua Xie
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chenyun Wang
- The Fourth Clinical Center Affiliated to Chinese PLA General Hospital, Beijing 100048, China
| | - Jing Dai
- Information Department, General Hospital of Western War Zone, Chengdu 610083, China
| | - Yunbo Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Hebei University, Baoding 071002, China.
| | - Lina Du
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Hebei University, Baoding 071002, China; Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
6
|
Wang Y, Gao J, Sun L, Li Q, Kang N, Gao C, Li T. Jia-Wei-Si-Miao-Yong-An Fang stimulates the healing of acute radiation-induced cutaneous wounds through MAPK/ERK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 306:116180. [PMID: 36693549 DOI: 10.1016/j.jep.2023.116180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE A famous traditional oral Chinese medicine formula, Si-Miao-Yong-An decoction, has been used to treat thromboangiitis obliterans from the Qing Dynasty. Because its therapeutic principles including clearing away heat, detoxification, accelerating blood circulation and relieving pains are consistent with acute radiation-induced cutaneous wounds in traditional Chinese medicine, we tried to add herbs and improve them into an external dosage form, called Jia-Wei-Si-Miao-Yong-An Fang (JWSMYA). However, its mechanism on radiation-induced cutaneous wounds is still unknown. AIM OF THE STUDY This study evaluated the therapeutic effect of JWSMYA and investigated the mechanism of repair and anti-fibrosis on acute radiation-induced cutaneous wounds with JWSMYA. MATERIALS AND METHODS Firstly, we prepared JWSMYA, and determined the composition through UHPLC LC-MS/MS. Then we used ionizing radiation to make a cutaneous wound model of rats, and observed wound healing through their skin injury score, wound contraction percentage and histological staining. In addition, immunohistochemical staining, Western blot analysis, qRT-PCR and Elisa were used to explore wound rehabilitation and anti-fibrosis mechanisms. RESULTS An in vivo assay revealed that JWSMYA promoted the repairment of acute radiation-induced cutaneous wounds, facilitated MAPK/ERK phosphorylation, inhibited PI3K/AKT activation, reduced the level of alpha-smooth muscle actin (a-sma), collagen type-I alpha 2 (Col1a2) and transforming growth factor-beta 1 (TGF-β1) in cutaneous tissues. However, no statistical difference was found in vascular endothelial growth factor (VEGF). CONCLUSION JWSMYA accelerated the repair of acute radiation-induced cutaneous wounds, which might be associated with the MAPK/ERK pathway. In addition, PI3K/AKT might be associated with the inhibition of fibrosis and the promotion of high-quality wound healing.
Collapse
Affiliation(s)
- Yin Wang
- Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Junfeng Gao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100071, People's Republic of China
| | - Liqiao Sun
- Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Qi Li
- Department of Oncology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Ning Kang
- Department of Oncology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Chen Gao
- Department of Oncology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Tong Li
- Department of Oncology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.
| |
Collapse
|
7
|
Muacevic A, Adler JR, Torres R, Maita K, Garcia J, Serrano L, Ho O, Forte AJ. Modulation of Burn Hypermetabolism in Preclinical Models. Cureus 2023; 15:e33518. [PMID: 36779088 PMCID: PMC9904913 DOI: 10.7759/cureus.33518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2023] [Indexed: 01/11/2023] Open
Abstract
Severe burns elicit a state of physiological stress and increased metabolism to help the body compensate for the changes associated with the traumatic injury. However, this hypermetabolic state is associated with increased insulin resistance, cardiovascular dysfunction, skeletal muscle catabolism, impaired wound healing, and delayed recovery. Several interventions were attempted to modulate burn hypermetabolism, including nutritional support, early excision and grafting, and growth hormone application. However, burn hypermetabolism still imposes significant morbidity and mortality in burn patients. Due to the limitations of in vitro models, animal models are indispensable in burn research. Animal models provide researchers with invaluable tools to test the safety and efficacy of novel treatments or advance our knowledge of previously utilized agents. Several animal studies evaluated novel therapies to modulate burn hypermetabolism in the last few years, including recombinant human growth hormone, erythropoietin, acipimox, apelin, anti-interleukin-6 monoclonal antibody, and ghrelin therapies. Results from these studies are promising and may be effectively translated into human studies. In addition, other studies revisited drugs previously used in clinical practice, such as insulin and metformin, to further investigate their underlying mechanisms as modulators of burn hypermetabolism. This review aims to update burn experts with the novel therapies under investigation in burn hypermetabolism with a focus on applicability and translation. Furthermore, we aim to guide researchers in selecting the correct animal model for their experiments by providing a summary of the methodology and the rationale of the latest studies.
Collapse
|
8
|
Zhang L, Yang R, Hu Y, Yang Y, Zhang X, He B, Shen Z, Yang J, Chen P. Promoting effect of pomegranate peel extract on second-degree burn wound-healing through VEGF-A and TGF-β1 regulation. Burns 2022; 48:639-648. [PMID: 34716043 DOI: 10.1016/j.burns.2021.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/13/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022]
Abstract
Pomegranate peel extract (PPE), a polyphenolic compound derived from pomegranate, has been widely concerned for its anti-oxidant, anti-inflammatory, and bacteriostatic effects. The potential therapeutic effect of PPE on burn injury was investigated, and its possible mechanisms were explored. Minipigs with second-degree burn were treated with PPE, Jing Wan Hong, and silver sulfadiazine. Hematoxylin-eosin (HE) staining was performed to detect burn severity, and then biological tissues were biopsied on days 0, 7, 14, 21, and 28 after administration. Immunohistochemistry, western blot, and real-time polymerase chain reaction (RT-PCR) were used to detect the protein and mRNA expression levels of VEGF-A and TGF-β1 in skin tissues after treatment with PPE. Furthermore, the skin wound healing at different time points was monitored by macroscopic observation. HE showed that after 28-day PPE treatment, the morphology of the skin tissue showed a significant improvement. Macroscopic data monitoring indicated that the decrustation and fur growing time was shortened. Meanwhile, the rate of wound healing increased after PPE treatment. The combination of immunohistochemistry, western blotting, and RT-PCR showed that after PPE treatment, expression of VEGF-A and TGF-β1 increased sharply on day 7, maintaining a high level until day 14, showing a downward trend on day 21, and approaching normal levels on day 28. However, in the model group, the protein and mRNA expression levels of VEGF-A and TGF-β1 increased on day 28 after burn injury, which was a slow process. Results indicated that compared with the model group, the peak expression level of VEGF-A and TGF-β1 was earlier, which was consistent with decrustation, shortening of fur growing time, and improvement of wound healing rate in minipig second-degree burn model. PPE showed a significant promoting effect on minipig second-degree burn model, which might be associated with the upregulation of the protein and gene expression levels of VEGF-A and TGF-β1.
Collapse
Affiliation(s)
- Li Zhang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China
| | - Renhua Yang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China
| | - Yangyang Hu
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China
| | - Yan Yang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China
| | - Xiaochao Zhang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China
| | - Bo He
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China
| | - Zhiqiang Shen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China
| | - JianYu Yang
- School of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China.
| | - Peng Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan 650500, China.
| |
Collapse
|
9
|
Jeong H, Chong HJ, So J, Jo Y, Yune TY, Ju BG. Ghrelin Represses Thymic Stromal Lymphopoietin Gene Expression through Activation of Glucocorticoid Receptor and Protein Kinase C Delta in Inflamed Skin Keratinocytes. Int J Mol Sci 2022; 23:ijms23073977. [PMID: 35409338 PMCID: PMC8999772 DOI: 10.3390/ijms23073977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 12/04/2022] Open
Abstract
Ghrelin, a peptide hormone secreted from enteroendocrine cells of the gastrointestinal tract, has anti-inflammatory activity in skin diseases, including dermatitis and psoriasis. However, the molecular mechanism underlying the beneficial effect of ghrelin on skin inflammation is not clear. In this study, we found that ghrelin alleviates atopic dermatitis (AD)-phenotypes through suppression of thymic stromal lymphopoietin (TSLP) gene activation. Knockdown or antagonist treatment of growth hormone secretagogue receptor 1a (GHSR1a), the receptor for ghrelin, suppressed ghrelin-induced alleviation of AD-like phenotypes and suppression of TSLP gene activation. We further found that ghrelin induces activation of the glucocorticoid receptor (GR), leading to the binding of GR with histone deacetylase 3 (HDAC3) and nuclear receptor corepressor (NCoR) NCoR corepressor to negative glucocorticoid response element (nGRE) on the TSLP gene promoter. In addition, ghrelin-induced protein kinase C δ (PKCδ)-mediated phosphorylation of p300 at serine 89 (S89), which decreased the acetylation and DNA binding activity of nuclear factor- κB (NF-κB) p65 to the TSLP gene promoter. Knockdown of PKCδ abolished ghrelin-induced suppression of TSLP gene activation. Our study suggests that ghrelin may help to reduce skin inflammation through GR and PKCδ-p300-NF-κB-mediated suppression of TSLP gene activation.
Collapse
Affiliation(s)
- Hayan Jeong
- Department of Life Science, Sogang University, Seoul 04107, Korea; (H.J.); (H.-J.C.); (J.S.); (Y.J.)
| | - Hyo-Jin Chong
- Department of Life Science, Sogang University, Seoul 04107, Korea; (H.J.); (H.-J.C.); (J.S.); (Y.J.)
| | - Jangho So
- Department of Life Science, Sogang University, Seoul 04107, Korea; (H.J.); (H.-J.C.); (J.S.); (Y.J.)
| | - Yejin Jo
- Department of Life Science, Sogang University, Seoul 04107, Korea; (H.J.); (H.-J.C.); (J.S.); (Y.J.)
| | - Tae-Young Yune
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul 02447, Korea;
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Bong-Gun Ju
- Department of Life Science, Sogang University, Seoul 04107, Korea; (H.J.); (H.-J.C.); (J.S.); (Y.J.)
- Correspondence: ; Tel.: +82-2-705-8455
| |
Collapse
|
10
|
Effect of Bacterial Infection on Ghrelin Receptor Regulation in Periodontal Cells and Tissues. Int J Mol Sci 2022; 23:ijms23063039. [PMID: 35328456 PMCID: PMC8950409 DOI: 10.3390/ijms23063039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
The effect of bacterial infection on the expression of growth hormone secretagogue receptor (GHS-R) was investigated in periodontal cells and tissues, and the actions of ghrelin were evaluated. GHS-R was assessed in periodontal tissues of rats with and without periodontitis. Human gingival fibroblasts (HGFs) were exposed to Fusobacterium nucleatum in the presence and absence of ghrelin. GHS-R expression was determined by real-time PCR and immunocytochemistry. Furthermore, wound healing, cell viability, proliferation, and migration were evaluated. GHS-R expression was significantly higher at periodontitis sites as compared to healthy sites in rat tissues. F. nucleatum significantly increased the GHS-R expression and protein level in HGFs. Moreover, ghrelin significantly abrogated the stimulatory effects of F. nucleatum on CCL2 and IL-6 expressions in HGFs and did not affect cell viability and proliferation significantly. Ghrelin stimulated while F. nucleatum decreased wound closure, probably due to reduced cell migration. Our results show original evidence that bacterial infection upregulates GHS-R in rat periodontal tissues and HGFs. Moreover, our study shows that ghrelin inhibited the proinflammatory actions of F. nucleatum on HGFs without interfering with cell viability and proliferation, suggesting that ghrelin and its receptor may act as a protective molecule during bacterial infection on periodontal cells.
Collapse
|
11
|
Zhang J, Zhu Y, Zhang Y, Lin W, Ke J, Liu J, Zhang L, Liu J. A balanced charged hydrogel with anti-biofouling and antioxidant properties for treatment of irradiation-induced skin injury. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112538. [PMID: 34857314 DOI: 10.1016/j.msec.2021.112538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 01/18/2023]
Abstract
Skin injury caused by large doses of ionizing radiation is the common and severe side effect of radiotherapy. However, its therapeutic efficacy is always hindered by early reactive oxygen species generation, repetitive inflammatory microenvironment and bacterial infection risk. Herein, we report an anti-biofouling hydrogel with anti-inflammation and anti-oxidative properties for the treatment of irradiation-induced skin injury. The anti-biofouling hydrogel can be achieved by balancing oppositely charged alginate, hyaluronic acid (HA) and polylysine (PLL) at the optimal ratio, which effectively resist protein and bacterial adhesion, and evades immune response. Moreover, curcumin and epigallocatechin gallate (EGCG) can be facially encapsulated and substantially released from the hydrogel. Results showed that the resulting AHP-Cur/EGCG hydrogel can significantly weaken the development of skin injury and accelerate its healing process by alleviating inflammation, scavenging ROS and promoting angiogenesis. Therefore, the findings presented in this work provide an effective strategy for clinical management and treatment of ionizing radiation-induced skin injury.
Collapse
Affiliation(s)
- Jiamin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yingnan Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yumin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Wenjing Lin
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Jia Ke
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin 300350, China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Lei Zhang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin 300350, China.
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
12
|
Peptides from Animal Origin: A Systematic Review on Biological Sources and Effects on Skin Wounds. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4352761. [PMID: 33149808 PMCID: PMC7603624 DOI: 10.1155/2020/4352761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/02/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022]
Abstract
Background Skin wounds are closely correlated with opportunistic infections and sepsis risk. Due to the need of more efficient healing drugs, animal peptides are emerging as new molecular platforms to accelerate skin wound closure and to prevent and control bacterial infection. Aim The aim of this study was to evaluate the preclinical evidence on the impact of animal peptides on skin wound healing. In addition, we carried out a critical analysis of the studies' methodological quality. Main Methods. This systematic review was performed according to the PRISMA guidelines, using a structured search on the PubMed-Medline, Scopus, and Web of Science platforms to retrieve studies published until August 25, 2020 at 3 : 00 pm. The studies included were limited to those that used animal models, investigated the effect of animal peptides with no association with other compounds on wound healing, and that were published in English. Bias analysis and methodological quality assessments were examined through the SYRCLE's RoB tool. Results Thirty studies were identified using the PRISMA workflow. In general, animal peptides were effective in accelerating skin wound healing, especially by increasing cellular proliferation, neoangiogenesis, colagenogenesis, and reepithelialization. Considering standardized methodological quality indicators, we identified a marked heterogeneity in research protocols and a high risk of bias associated with limited characterization of the experimental designs. Conclusion Animal peptides show a remarkable healing potential with biotechnological relevance for regenerative medicine. However, rigorous experimental approaches are still required to clearly delimit the mechanisms underlying the healing effects and the risk-benefit ratio attributed to peptide-based treatments.
Collapse
|
13
|
Myung H, Jang H, Myung JK, Lee C, Lee J, Kang J, Jang WS, Lee SJ, Kim H, Kim HY, Park S, Shim S. Platelet-rich plasma improves the therapeutic efficacy of mesenchymal stem cells by enhancing their secretion of angiogenic factors in a combined radiation and wound injury model. Exp Dermatol 2019; 29:158-167. [PMID: 31560791 DOI: 10.1111/exd.14042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/18/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022]
Abstract
Delayed wound healing after radiation exposure can cause serious cutaneous damage, and its treatment is a major clinical challenge. Although mesenchymal stem cells (MSCs) have emerged as a promising therapeutic agent in regenerative medicine, they alone do not produce satisfactory effects in a combined radiation and wound injury (CRWI) model. Here, we investigated the therapeutic effect of combined umbilical cord blood-derived (UCB)-MSCs and platelet-rich plasma (PRP) treatment on wound healing in a CRWI mouse model. First, we assessed the release of cytokines from UCB-MSCs cultured with PRP and observed changes in the expression of angiogenic factors. The angiogenic paracrine factors from UCB-MSCs cultured with PRP were assessed in human umbilical vein endothelial cells (HUVECs). To assess therapeutic efficacy, UCB-MSCs and PRP were topically implanted into a CRWT mouse model. Vascular endothelial growth factor (VEGF), a pro-angiogenic growth factor, urokinase-type plasminogen activator and contributor to VEGF-induced signalling were more highly expressed in conditioned media of UCB-MSCs cultured with PRP than in that of UCB-MSCs alone. Furthermore, conditioned media of UCB-MSCs cultured with PRP increased the formation of tube-like structures in HUVECs. Co-treatment of UCB-MSCs and PRP in a CRWI mouse model increased the wound closure rate and angiogenesis compared with an untreated irradiated group. Moreover, increased expression of VEGF and CD31 were observed in the wound tissue of co-treated mice compared with untreated irradiated mice. PRP stimulates the release of angiogenic factors from UCB-MSCs, and combined therapy of UCB-MSCs and PRP improves regeneration efficacy by enhancing angiogenesis in a CRWI model.
Collapse
Affiliation(s)
- Hyunwook Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.,Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Changsun Lee
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Janet Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - JiHoon Kang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hwi-Yool Kim
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| |
Collapse
|
14
|
Zhang K, Zhang Y, Li N, Xing F, Zhao J, Yang T, Liu C, Feng N. An herbal-compound-based combination therapy that relieves cirrhotic ascites by affecting the L-arginine/nitric oxide pathway: A metabolomics-based systematic study. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:112034. [PMID: 31226385 DOI: 10.1016/j.jep.2019.112034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/30/2019] [Accepted: 06/17/2019] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine boasts a 440-year-long history of treating refractory ascites via combinations of herbal medicines, called formulae. Xiaozhang Tie (XT) is a proprietary herbal-compound-based formula that has been proven to be very effective in the treatment of cirrhosis-associated ascites in clinical practice, but the mechanism of action of XT remains unknown. AIM OF THE STUDY In this study, we used a metabolomics-based systematic method to elucidate the mechanism of XT in the treatment of cirrhotic ascites. METHODS Decompensated liver cirrhosis was induced in rats by intraperitoneal injection of Carbon tetrachloride (CCl4). Ultra performance liquid chromatography-mass spectrometry (UPLC-MS) combined with pattern recognition approaches were used to determine differentiating metabolites relevant to XT treatment. Biomarkers were further validated by a targeted quantitative method and by the results from serum and urine analyses. Pathway analysis and correlation network construction were used to reveal the therapeutic targets associated with XT treatment, and the potential mechanisms were verified by the results from biochemical, histopathological and immunohistochemical assays. RESULTS XT synergistically mediated the abnormalities of amino acid metabolic pathways in cirrhotic rats. XT significantly elevated the arginine levels, reduced the serum nitric oxide (NO) levels and alleviated the gastrointestinal motility disorder of cirrhotic rats. This effect of XT has been confirmed by the inhibition of the activities of inducible NO synthase and neuronal NO synthase in the small intestine. CONCLUSIONS These results reveal that XT promotes gastrointestinal motility by acting on multiple targets in multiple pathways, of which the L-arginine/NO pathway is most affected.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China; First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| | - Yongtai Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Nana Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| | - Feng Xing
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, People's Republic of China.
| | - Jihui Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| | - Tao Yang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, People's Republic of China.
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, People's Republic of China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
15
|
Lyra Junior HF, de Lucca Schiavon L, Rodrigues IK, Couto Vieira DS, de Paula Martins R, Turnes BL, Latini AS, D'Acâmpora AJ. Effects of Ghrelin on the Oxidative Stress and Healing of the Colonic Anastomosis in Rats. J Surg Res 2018; 234:167-177. [PMID: 30527470 DOI: 10.1016/j.jss.2018.09.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Anastomotic leakage is the deadliest complication of colonic procedures. Ghrelin is an orexigenic hormone with potent actions on growth hormone release and functions in the processes of growth, tissue inflammation, repair, and oxidative stress. We evaluated the hypothesis that the exogenous administration of ghrelin causes beneficial effects on the healing of colonic anastomosis. MATERIALS AND METHODS Sixty-four male Wistar rats were randomly assigned to eight subgroups receiving postoperative intraperitoneal administration of ghrelin (23 μg/kg/d) or saline after a colonic anastomosis. The anastomotic tissue was evaluated on the third, seventh, and 14th postoperative days. Anastomotic bursting pressure, histological parameters, hydroxyproline content, and tissue oxidative stress markers were compared. RESULTS There was a significant increase in the mean anastomotic bursting pressure in the ghrelin subgroup on the seventh postoperative day (P = 0.035). Histological evaluation demonstrated a significant difference in the neutrophilic infiltrate (P = 0.035) on the third and 14th d and in apoptosis (P = 0.004), granulation tissue (P = 0.011) and peritoneal inflammation (P = 0.014) on the 14th postoperative day. There was a statistically significant increase in the hydroxyproline content in the ghrelin subgroup on the 14th postoperative day (P = 0.043). There were significant differences in the nitrite tissue levels (P = 0.021) on day 3 and in reactive oxygen species (P = 0.012) on day 14. CONCLUSIONS The administration of ghrelin had beneficial anti-inflammatory and antioxidant effects, increasing the resistance of the anastomosis and the hydroxyproline tissue content in the postoperative period.
Collapse
Affiliation(s)
| | - Leonardo de Lucca Schiavon
- Department of Internal Medicine, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Igor Kunze Rodrigues
- Department of Surgery, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | | | - Roberta de Paula Martins
- Department of Biochemistry, Bioenergetics and Oxidative Stress Laboratory, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Bruna Lenfers Turnes
- Department of Biochemistry, Bioenergetics and Oxidative Stress Laboratory, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Alexandra Susana Latini
- Department of Biochemistry, Bioenergetics and Oxidative Stress Laboratory, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Armando José D'Acâmpora
- Department of Surgery, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| |
Collapse
|
16
|
Eming SA, Tomic-Canic M. Updates in wound healing: Mechanisms and translation. Exp Dermatol 2018; 26:97-98. [PMID: 28133858 DOI: 10.1111/exd.13281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Sabine A Eming
- Department of Dermatology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Marjana Tomic-Canic
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
17
|
Asprosin: Possible target in connection with ghrelin and cytokine network expression in the post-burn treatment. Med Hypotheses 2018; 118:163-168. [DOI: 10.1016/j.mehy.2018.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/29/2018] [Accepted: 07/05/2018] [Indexed: 12/14/2022]
|
18
|
Brzozowska A, Homa-Mlak I, Mlak R, Gołębiowski P, Mazurek M, Ciesielka M, Małecka-Massalska T. Polymorphism of regulatory region of GHRL gene (-2531C>T) as a promising predictive factor for radiotherapy-induced oral mucositis in patients with head neck cancer. Head Neck 2018; 40:1799-1811. [PMID: 29566446 DOI: 10.1002/hed.25154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/22/2017] [Accepted: 02/08/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The purpose of this study was to investigate the relationship between single nucleotide polymorphisms (SNP; rs1629816) in the regulatory region (c.-2531C>T) of the ghrelin (GHRL) gene and the occurrence and severity of oral mucositis caused by radiotherapy (RT) in patients with head and neck cancer. METHODS Oral mucositis in 65 patients with head and neck cancer who underwent irradiation were assessed according to Radiation Therapy Oncology Group (RTOG)/European Organisation for Research and Treatment of Cancer (EORTC) scale. The DNA from patients with head and neck cancer was isolated from whole blood. The genotypes were determined using the minisequencing method (SNaPshot PCR). RESULTS The frequency of occurrence of the GHRL gene (c.-2531C>T, rs1629816) genotypes were as follows: AA = 21.5%; GA = 40%; and GG = 38.5%. In case of AA genotype, there was a 7-fold decrease of the risk of occurrence of oral mucositis (of grades 2 and 3) in the sixth week of RT (AA vs GA or GG, respectively: 17.9% vs 82.1% patients; odds ratio [OR] 0.14; 95% confidence interval [CI] 0.02-0.98; P = .0481). No statistically significant differences were observed between the volume of oral cavity contours (V30, V40, and V50) depending on the GHRL genotype in patients with head and neck cancer. CONCLUSION The study results have demonstrated an association between the AA genotype of the GHRL gene and the risk of more severe oral mucositis attributed to RT in patients with head and neck cancer.
Collapse
Affiliation(s)
- Anna Brzozowska
- Department of Oncology, Medical University of Lublin, Lublin, Poland
| | - Iwona Homa-Mlak
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland
| | - Radosław Mlak
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland
| | - Paweł Gołębiowski
- Department of Oncology, Medical University of Lublin, Lublin, Poland
| | - Marcin Mazurek
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland
| | - Marzanna Ciesielka
- Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
19
|
Ghrelin protects against depleted uranium-induced bone damage by increasing osteoprotegerin/RANKL ratio. Toxicol Appl Pharmacol 2018; 343:62-70. [DOI: 10.1016/j.taap.2018.02.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/10/2018] [Accepted: 02/21/2018] [Indexed: 01/03/2023]
|
20
|
Ye M, Huang R, Min Z, Zhang P, Wang T, Yu B. Comparison of the effect by which gastric plication and sleeve gastrectomy procedures alter metabolic and physical parameters in an obese type 2 diabetes rodent model. Surg Obes Relat Dis 2017. [DOI: 10.1016/j.soard.2017.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Li W, Wu X, Qu R, Wang W, Chen X, Cheng L, Liu Y, Guo L, Zhao Y, Liu C. Ghrelin protects against nucleus pulposus degeneration through inhibition of NF-κB signaling pathway and activation of Akt signaling pathway. Oncotarget 2017; 8:91887-91901. [PMID: 29190883 PMCID: PMC5696149 DOI: 10.18632/oncotarget.19695] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
The objective of the present study was to examine the potential role of ghrelin in degeneration of nucleus pulposus (NP). Lower expression levels of ghrelin were found in human NP cells stimulated with interleukin-1β (IL-1β). Moreover, exogenous ghrelin suppressed IL-1β induced degeneration and inflammation associated biomarkers in human NP cells, including matrix metalloproteinase-13, a disintegrin and metalloproteinase with thrombospondin motifs-5, tumor necrosis factor-α and iNOS, which was possibly mediated by antagonization of NF-κB signaling. Moreover, ghrelin enhanced production of critical extracellular matrix of NP cells, including collagen 2, aggrecan, and Sox-9 in NP cells. Ghrelin also promoted NP tissue regeneration in a rabbit IVD degeneration model, which seems to be associated with growth hormone secretagogue receptor. Additionally, the protective role of ghrelin in anabolism potentially relies on activation of Akt signaling pathway. Taken together, ghrelin may represent a molecular target for prevention and treatment of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P. R. China
| | - Xihai Wu
- Department of Gynaecology and Obstetrics, Jinan Central Hospital, Shandong University, Jinan, Shandong 250012, P. R. China
| | - Ruize Qu
- Medical School of Shandong University, Jinan, Shandong 250012, P. R. China
| | - Wenhan Wang
- Medical School of Shandong University, Jinan, Shandong 250012, P. R. China
| | - Xiaomin Chen
- Medical School of Shandong University, Jinan, Shandong 250012, P. R. China
| | - Lei Cheng
- Department of Orthopaedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P. R. China
| | - Yaoge Liu
- Medical School of Shandong University, Jinan, Shandong 250012, P. R. China
| | - Linlin Guo
- Medical School of Shandong University, Jinan, Shandong 250012, P. R. China
| | - Yunpeng Zhao
- Department of Orthopaedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P. R. China
| | - Chao Liu
- Department of Oral and Maxillofacial Surgery and Institute of Dental Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P. R. China
| |
Collapse
|