Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Artigo em Chinês | MEDLINE | ID: mdl-36878519

RESUMO

Wound healing is a complex process under precise regulation, including multiple stages such as inflammation, anti-inflammatory, and regeneration. Macrophages play an important regulatory role in the differentiated process of wound healing due to their obvious plasticity. If macrophages fail to express specific functions in a timely manner, it will affect the healing function of tissues and lead to pathological tissue healing. Therefore, it is of great significance to understand the different functions of different types of macrophages and to regulate them specifically in different stages of wound healing to promote the healing and regeneration of wound tissue. In this paper, we illustrate the different functions of macrophages in the wound and their basic mechanisms, according to the basic process of wound healing, and emphasize the strategies of macrophage regulation that may be applied to clinical treatment in the future.


Assuntos
Inflamação , Macrófagos , Humanos , Cicatrização
2.
Zhonghua Shao Shang Za Zhi ; 38(9): 830-838, 2022 Sep 20.
Artigo em Chinês | MEDLINE | ID: mdl-36177587

RESUMO

Objective: To investigate the effects of low-dose photodynamic therapy on the proliferation, regulation, and secretion functions of human adipose mesenchymal stem cells (ADSCs) and the related mechanism, so as to explore a new method for the repair of chronic wounds. Methods: The experimental research methods were adopted. From February to April 2021, 10 patients (5 males and 5 females, aged 23 to 47 years) who underwent cutaneous surgery in the Department of Dermatology of the First Affiliated Hospital of Army Medical University (the Third Military Medical University) donated postoperative waste adipose tissue. The cells were extracted from the adipose tissue and the phenotype was identified. Three batches of ADSCs were taken, with each batch of cells being divided into normal control group with conventional culture only, photosensitizer alone group with conventional culture after being treated with Hemoporfin, irradiation alone group with conventional culture after being treated with red light irradiation, and photosensitizer+irradiation group with conventional culture after being treated with Hemoporfin and red light irradiation, with sample number of 3 in each group. At culture hour of 24 after the treatment of the first and second batches of cells, the ADSC proliferation level was evaluated by 5-ethynyl-2'-deoxyuridine staining method and the migration percentage of HaCaT cells cocultured with ADSCs was detected by Transwell experiment, respectively. On culture day of 7 after the treatment of the third batch of cells, the extracellular matrix protein expression of ADSCs was detected by immunofluorescence method. The ADSCs were divided into 0 min post-photodynamic therapy group, 15 min post-photodynamic therapy group, 30 min post-photodynamic therapy group, and 60 min post-photodynamic therapy group, with 3 wells in each group. Western blotting was used to detect the protein expressions and calculate the phosphorylated mammalian target of rapamycin complex (p-mTOR)/mammalian target of rapamycin (mTOR), phosphorylated p70 ribosomal protein S6 kinase (p-p70 S6K)/p70 ribosomal protein S6 kinase (p70 S6K) ratio at the corresponding time points after photodynamic therapy. Two batches of ADSCs were taken, and each batch was divided into normal control group, photodynamic therapy alone group, and photodynamic therapy+rapamycin group, with 3 wells in each group. At culture minute of 15 after the treatment, p-mTOR/mTOR and p-p70 S6K/p70 S6K ratios of cells from the first batch were calculated and detected as before. On culture day of 7 after the treatment, extracellular matrix protein expression of cells from the second batch was detected as before. Data were statistically analyzed with one-way analysis of variance and least significant difference test. Results: After 12 d of culture, the cells were verified as ADSCs. At culture hour of 24 after the treatment, the ADSC proliferation level ((4.0±1.0)% and (4.1±0.4)%, respectively) and HaCaT cell migration percentages (1.17±0.14 and 1.13±0.12, respectively) in photosensitizer alone group and irradiation alone group were similar to those of normal control group ((3.7±0.6)% and 1.00±0.16, respectively, P>0.05), and were significantly lower than those of photosensitizer+irradiation group ((34.2±7.0)% and 2.55±0.13, respectively, P<0.01). On culture day of 7 after the treatment, compared with those in normal control group, the expression of collagen Ⅲ in ADSCs of photosensitizer alone group was significantly increased (P<0.05), and the expressions of collagen Ⅰ and collagen Ⅲ in ADSCs of irradiation alone group were significantly increased (P<0.01). Compared with those in photosensitizer alone group and irradiation alone group, the expressions of collagen Ⅰ, collagen Ⅲ, and fibronectin of ADSCs in photosensitizer+irradiation group were significantly increased (P<0.01). Compared with those in 0 min post-photodynamic therapy group, the ratios of p-mTOR/mTOR and p-p70 S6K/p70 S6K of ADSCs in 15 min post-photodynamic therapy group were significantly increased (P<0.01), the ratios of p-p70 S6K/p70 S6K of ADSCs in 30 min post-photodynamic therapy group and 60 min post-photodynamic therapy group were both significantly increased (P<0.01). At culture minute of 15 after the treatment, compared with those in normal control group, the ratios of p-mTOR/mTOR and p-p70 S6K/p70 S6K of ADSCs in photodynamic therapy alone group were significantly increased (P<0.05 or P<0.01). Compared with those in photodynamic therapy alone group, the ratios of p-mTOR/mTOR and p-p70 S6K/p70 S6K of ADSCs in photodynamic therapy+rapamycin group were significantly decreased (P<0.05). On culture day of 7 after the treatment, compared with those in normal control group, the expressions of collagen Ⅰ, collagen Ⅲ, and fibronectin of ADSCs in photodynamic therapy alone group were significantly increased (P<0.01). Compared with those in photodynamic therapy alone group, the expressions of collagen Ⅰ, collagen Ⅲ, and fibronectin of ADSCs in photodynamic therapy+rapamycin group were significantly decreased (P<0.01). Conclusions: Low-dose photodynamic therapy can promote the proliferation of ADSCs, improve the ability of ADSCs to regulate the migration of HaCaT cells, and enhance the secretion of extracellular matrix protein by rapidly activating mTOR signaling pathway.


Assuntos
Células-Tronco Mesenquimais , Fotoquimioterapia , Tecido Adiposo , Feminino , Fibronectinas , Humanos , Masculino , Fármacos Fotossensibilizantes/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
3.
Zhonghua Shao Shang Za Zhi ; 38(9): 897-898, 2022 Sep 20.
Artigo em Chinês | MEDLINE | ID: mdl-36177599

RESUMO

The 17th Chinese Symposium on Burn Medicine and the 2022 Congress of Burn Medicine Branch of China International Exchange and Promotion Association for Medical and Healthcare, and the 12th Academic Conference on Burn and Plastic Surgery in Five Provinces and One City in Southwest China was successfully held in green city Nanning, from August 25th to 27th, 2022. The conference theme was "Burn treatment and wound repair", received nearly 200 submissions, nearly 1 100 online and offline registered delegates, and nearly 300 offline attendees. The meetings were held in one main venue and three branch venues, with combination of speaking offline and live and recorded broadcast, as well as whole process synchronous live broadcasting. During the meeting, key issues about burn treatment and wound repair were discussed, with warm academic atmosphere.


Assuntos
Queimaduras , Procedimentos de Cirurgia Plástica , Cirurgia Plástica , Queimaduras/cirurgia , China , Atenção à Saúde , Humanos
4.
Zhonghua Shao Shang Za Zhi ; 38(2): 114-118, 2022 Feb 20.
Artigo em Chinês | MEDLINE | ID: mdl-35220699

RESUMO

Re-epithelialization is one of the core links that determines the healing process of skin wounds. The proliferation and differentiation of epidermal stem cells to form new epidermal tissue is the histological basis of re-epithelialization, and the smooth progress of the cell differentiation process of epidermal stem cells-precursor cells-terminal cells is the cytological basis for the continuous formation of new epidermal tissue. The proliferation of stem cells and their differentiation into precursor cells are the determinants of the proliferative potential of newly formed epidermal tissue, while the expansion and differentiation of precursor cells into terminal cells are key factors determining the rate of new epidermal tissue formation. The tissue microenvironment plays a key regulatory role in the process of wound re-epithelialization, and cell growth factor and inflammatory mediators are the two main components of tissue microenvironment, which play regulatory role in different aspects of proliferation and differentiation of epidermal stem cells, jointly promoting the smooth progress of wound re-epithelialization As an important part of skin immune system, the subsets of gamma-delta (γδ) T cells play crucial role in dynamically shaping early wound microenvironment via secreting different cell growth factors and inflammatory factors. From the prospective of immune microenvironment of wound, this paper discusses the role of skin γδ T cells in maintaining the balance of stem cell proliferation and differentiation and regulating wound re-epithelialization, providing a new direction for the prevention and treatment of refractory wound.


Assuntos
Reepitelização , Linfócitos T , Estudos Prospectivos , Pele , Subpopulações de Linfócitos T
5.
Zhonghua Shao Shang Za Zhi ; 38(2): 119-129, 2022 Feb 20.
Artigo em Chinês | MEDLINE | ID: mdl-35220700

RESUMO

Objective: To explore the effects of P311 on the angiogenesis ability of human microvascular endothelial cell 1 (HMEC-1) in vitro and the potential molecular mechanism. Methods: The experimental research method was used. HMEC-1 was collected and divided into P311 adenovirus group and empty adenovirus group according to the random number table (the same grouping method below), which were transfected correspondingly for 48 h. The cell proliferation activity was detected using the cell counting kit 8 on 1, 3, and 5 days of culture. The residual scratch area of cells at post scratch hour 6 and 11 was detected by scratch test, and the percentage of the residual scratch area was calculated. The blood vessel formation of cells at 8 h of culture was observed by angiogenesis experiment in vitro, and the number of nodes and total length of the tubular structure were measured. The protein expressions of vascular endothelial growth factor receptor 2 (VEGFR2), phosphorylated VEGFR2 (p-VEGFR2), extracellular signal-regulated kinase 1/2 (ERK1/2), and phosphorylated ERK1/2 (p-ERK1/2) in cells were detected by Western blotting. HMEC-1 was collected and divided into P311 adenovirus+small interfering RNA (siRNA) negative control group, empty adenovirus+siRNA negative control group, P311 adenovirus+siRNA-VEGFR2 group, and empty adenovirus+siRNA-VEGFG2 group, which were treated correspondingly. The protein expressions of VEGFR2, p-VEGFR2, ERK1/2, and p-ERK1/2 in cells were detected by Western blotting at 24 h of transfection. The blood vessel formation of cells at 24 h of transfection was observed by angiogenesis experiment in vitro, and the number of nodes and total length of the tubular structure were measured. HMEC-1 was collected and divided into P311 adenovirus+dimethylsulfoxide (DMSO) group, empty adenovirus+DMSO group, P311 adenovirus+ERK1/2 inhibitor group, and empty adenovirus+ERK1/2 inhibitor group, which were treated correspondingly. The protein expressions of ERK1/2 and p-ERK1/2 in cells were detected by Western blotting at 2 h of treatment. The blood vessel formation of cells at 2 h of treatment was observed by angiogenesis experiment in vitro, and the number of nodes and total length of the tubular structure were measured. The sample number at each time point in each group was 6. Data were statistically analyzed with independent sample t test, analysis of variance for repeated measurement, one-way analysis of variance, and least significant difference test. Results: Compared with that of empty adenovirus group, the proliferation activity of cells in P311 adenovirus group did not show significant difference on 1, 3, and 5 days of culture (with t values of -0.23, -1.30, and -1.52, respectively, P>0.05). The residual scratch area percentages of cells in P311 adenovirus group were significantly reduced at post scratch hour 6 and 11 compared with those of empty adenovirus group (with t values of -2.47 and -2.62, respectively, P<0.05). At 8 h of culture, compared with those of empty adenovirus group, the number of nodes and total length of the tubular structure of cells in P311 adenovirus group were significantly increased (with t values of 4.49 and 4.78, respectively, P<0.01). At 48 h of transfection, compared with those of empty adenovirus group, the protein expressions of VEGFR2 and ERK1/2 of cells in P311 adenovirus group showed no obvious changes (P>0.05), and the protein expressions of p-VEGFR2 and p-ERK1/2 of cells in P311 adenovirus group were significantly increased (with t values of 17.27 and 16.08, P<0.01). At 24 h of transfection, the protein expressions of p-VEGFR2 and p-ERK1/2 of cells in P311 adenovirus+siRNA negative control group were significantly higher than those in empty adenovirus+siRNA negative control group (P<0.01). The protein expressions of VEGFR2, p-VEGFR2, and p-ERK1/2 of cells in P311 adenovirus+siRNA negative control group were significantly higher than those in P311 adenovirus+siRNA-VEGFR2 group (P<0.01). The protein expressions of VEGFR2 and p-ERK1/2 of cells in empty adenovirus+siRNA negative control group were significantly higher than those in empty adenovirus+siRNA-VEGFR2 group (P<0.05 or P<0.01). At 24 h of transfection, the number of nodes of the tubular structure in cells of P311 adenovirus+siRNA negative control group was 720±62, which was significantly more than 428±38 in empty adenovirus+siRNA negative control group and 364±57 in P311 adenovirus+siRNA-VEGFR2 group (with P values both <0.01). The total length of the tubular structure of cells in P311 adenovirus+siRNA negative control group was (21 241±1 139) µm, which was significantly longer than (17 005±1 156) µm in empty adenovirus+siRNA negative control group and (13 494±2 465) µm in P311 adenovirus+siRNA-VEGFR2 group (with P values both <0.01). The number of nodes of the tubular structure in cells of empty adenovirus+siRNA negative control group was significantly more than 310±75 in empty adenovirus+siRNA-VEGFR2 group (P<0.01), and the total length of the tubular structure of cells in empty adenovirus+siRNA negative control group was significantly longer than (11 600±2 776) µm in empty adenovirus+siRNA-VEGFR2 group (P<0.01). At 2 h of treatment, the protein expression of p-ERK1/2 of cells in P311 adenovirus+DMSO group was significantly higher than that in empty adenovirus+DMSO group and P311 adenovirus+ERK1/2 inhibitor group (with P values both <0.01), and the protein expression of p-ERK1/2 of cells in empty adenovirus+DMSO group was significantly higher than that in empty adenovirus+ERK1/2 inhibitor group (P<0.05). At 2 h of treatment, the number of nodes of the tubular structure in cells of P311 adenovirus+DMSO group was 726±72, which was significantly more than 421±39 in empty adenovirus+DMSO group and 365±41 in P311 adenovirus+ERK1/2 inhibitor group (with P values both <0.01). The total length of the tubular structure of cells in P311 adenovirus+DMSO group was (20 318±1 433) µm, which was significantly longer than (16 846±1 464) µm in empty adenovirus+DMSO group and (15 114±1 950) µm in P311 adenovirus+ERK1/2 inhibitor group (with P values both <0.01). The number of nodes of the tubular structure in cells of empty adenovirus+DMSO group was significantly more than 317±67 in empty adenovirus+ERK1/2 inhibitor group (P<0.01), and the total length of the tubular structure of cells in empty adenovirus+DMSO group was significantly longer than (13 188±2 306) µm in empty adenovirus+ERK1/2 inhibitor group (P<0.01). Conclusions: P311 can enhance the angiogenesis ability of HMEC-1 by activating the VEGFR2/ERK1/2 signaling pathway.


Assuntos
Células Endoteliais , Proteínas do Tecido Nervoso , Proteínas Oncogênicas , Fator A de Crescimento do Endotélio Vascular , Adenoviridae/genética , Linhagem Celular , Endotélio Vascular , Humanos , Neovascularização Fisiológica , Transdução de Sinais , Transfecção
6.
Zhonghua Shao Shang Za Zhi ; 37(6): 596-600, 2021 Jun 20.
Artigo em Chinês | MEDLINE | ID: mdl-34167287

RESUMO

The 16th Chinese Symposium on Burn Medicine and the 2021 Congress of Burn Medicine Branch of China International Exchange and Promotion Association for Medical and Healthcare (CPAM) and the 2021 International Summit Forum of Burns in Chongqing was successfully held in Chongqing, from May 19th to 21st in 2021. A total of more than 500 specialists and scholars across the country attended the meeting. The theme of this congress was "Burn Medicine: standardization and internationalization" . With the meetings being held in the one main venue and three branch venues and elite forum, the related hot topics and difficult problems were discussed warmly in multiple dimensions. During the conference, Founding Congress of 6th Editorial Committee of Chinese Journal of Burns, the Standing Committee and whole Committee of Chinese Burn Association, and the Congress of Burn Medicine Branch of CPAM were held in pragmatic and efficient manners.


Assuntos
Queimaduras , Queimaduras/terapia , China , Atenção à Saúde , Humanos , Padrões de Referência
7.
Zhonghua Shao Shang Za Zhi ; 37(3): 296-300, 2021 Mar 20.
Artigo em Chinês | MEDLINE | ID: mdl-33765727

RESUMO

Wound healing is a complex and critical process, which includes three stages: inflammation, proliferation, and remodeling. The epidermal cells are precisely regulated in this process. On one hand, keratinocytes around the wound edge migrate and proliferate to form a new basement membrane to cover the wound. On the other hand, the epidermal stem cells are activated with the proliferation and differentiation being enhanced, and the terminal differentiation and apoptosis being inhibited; and together with keratinocytes, epidermal stem cells promote the process of re-epithelialization under the regulation of various factors. In the epidermis, there is a group of resident T cell subsets, dendritic epidermal lymphocytes (DETCs) that play a key role in protecting the function of epidermal tissue. DETCs are activated after recognizing unknown antigens, the activated DETCs secret cytokines such as insulin-like growth factor Ⅰ, keratinocyte growth factor-1/2, granulocyte-macrophage colony stimulating factor, interferon-γ, and transforming growth factor-ß, which promote epidermal homeostasis and re-epithelialization by regulating the dynamic balance among keratinocytes migration, proliferation, and apoptosis, and the differentiation of epidermal stem cells around the wound edge. This article discusses the biological characteristics of DETCs and their roles in the maintenance of epidermal homeostasis and wound healing.


Assuntos
Epiderme , Cicatrização , Diferenciação Celular , Movimento Celular , Células Epidérmicas , Queratinócitos
8.
Zhonghua Xin Xue Guan Bing Za Zhi ; 49(1): 43-48, 2021 Jan 11.
Artigo em Chinês | MEDLINE | ID: mdl-33429485

RESUMO

Objective: The purpose of this study was to investigate the effects of CYP2C19 gene mutations on clopidogrel antiplatelet activity in the patients with coronary heart disease treated by percutaneous coronary intervention. Methods: Patients with coronary heart disease, who hospitalized in the Second Affiliated Hospital of Nanchang University from March 2011 to June 2019, and healthy individuals with matching genetic background, gender, and age as controls were included in this study. Basic clinical data were analyzed and blood samples of all research subjects were obtained for extraction of DNA, and Sanger first-generation sequencing method was used to detect CYP2C19 gene mutation from full exon and exon and intron junction. CYP2C19 gene variations in patients with coronary heart disease were compared with the 1000 Genomes Browse database and the sequencing results of healthy controls to determine whether the gene variation was a genetic mutation or a genetic polymorphism. After that, PolyPhen-2 prediction software was used to analyze the harmfulness of gene mutations to predict the effect of mutations on protein function. The same dose of CYP2C19 wild-type plasmid and the CYP2C19 gene mutant plasmids were transfected into human normal liver cells HL-7702. After transfection of 24 h, the expression of CYP2C19 protease in each group was detected. The liver S9 protein was incubated with clopidogrel, acted on platelets to detect the platelet aggregation rate and the activity of human vasodilator-activated phosphoprotein (VASP). Results: A total of 1 493 patients with coronary heart disease (59.36%) were enrolled, the average age was (64.5±10.4) years old, of which 1 129 were male (75.62%). Meanwhile, 1 022 healthy physical examination volunteers (40.64%) were enrolled, and the average age was (64.1±11.0) years old, of which 778 were male (76.13%). A total of 5 gene mutations of CYP2C19 gene were identified in 12 patients (0.80%), namely, 4 known mutations T130K (1 case), M136K (6 cases), N277K (3 cases), V472I (1 case) and one new mutation G27V (1 case), no corresponding gene mutation was found in healthy controls. It was found that T130K and M136K were probably damaging, G27V was possibly damaging, and N277K and V472I were benign mutations. In vitro, we demonstrated that the platelet aggregation rate of the M136K gene mutation group was 24.83% lower than that of the wild type (59.58% vs. 34.75%; P<0.05), and the phosphorylated VASP level was 23.0% higher than that of the wild type (1.0 vs. 1.23; P<0.05). However, the platelet aggregation rate and phosphorylated VASP level were similar between of G27V, T130K, N277K, V472I gene mutation groups and wild type group (P>0.05). Conclusions: In this study, 5 gene mutations are defined in patients with coronary heart disease, namely G27V, T130K, M136K, N277K, V472I. In vitro functional studies show that CYP2C19 gene mutation M136K, as a gain-of-function gene mutation, can enhance the activation of CYP2C19 enzyme on clopidogrel, thereby inhibiting the platelet aggregation rate.

9.
Zhonghua Shao Shang Za Zhi ; 36(10): 901-904, 2020 Oct 20.
Artigo em Chinês | MEDLINE | ID: mdl-33105941

RESUMO

The immune system of skin is consisted of different types of immune cells which distribute in epidermis and dermis. By adjusting the microenvironment of wound, these immune cells regulate the migration, proliferation, differentiation and other functions of epidermal cells, endothelial cells, and fibroblasts, thus taking part in re-epithelialization and formation of granulation tissue and having great influences on wound healing in the end. Acknowledging the interaction of these cells and identifying the targets that regulate the functions of immune cells will provide direction for improving the speed and quality of wound healing.


Assuntos
Células Endoteliais , Cicatrização , Movimento Celular , Fibroblastos , Tecido de Granulação , Humanos , Queratinócitos , Reepitelização , Pele
10.
Zhonghua Shao Shang Za Zhi ; 36(10): 915-922, 2020 Oct 20.
Artigo em Chinês | MEDLINE | ID: mdl-33105943

RESUMO

Objective: To analyze target genes of human platelet-rich plasma (PRP) in regulating and controlling human epidermal stem cells (ESCs). Methods: (1) The discarded foreskin tissues were collected from 6 male patients of the First Affiliated Hospital of Army Medical University after urological surgery. The patients aged 5 to 25 years with good health and without urinary system infection. Human ESCs were cultivated using quick attachment method, and were subjected to morphological observation and identification. Venous blood sample in the volume of 40 mL was collected from a female healthy volunteer (aged 29 years) of General Hospital of Southern Theater Command of PLA, and PRP was extracted by second centrifugation method. (2) The successfully cultured primary human ESCs were divided into control group and PRP group according to the random number table, with 3 wells in each group. The cells in control group were not specially treated. In PRP group, PRP was added to the ESC medium to achieve final volume fraction of 2.5% after the cells were adhered for 12 hours. RNA was extracted, and transcriptome sequencing and data analysis of human ESCs of two groups were performed using RNA sequencing technology. Using the false discovery rate less than 0.05 and the fold change more than or equal to 4 as the standard, the differentially expressed genes were screened by Dr. Tom data mining system. Gene ontology (GO) enrichment analysis was performed on the obtained differentially expressed genes to find out the GO entries with significant enrichment. Then Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation analysis was used to further analyze the biological processes or metabolic pathways in which differentially expressed genes might be involved. Finally, the genes related to re-epithelialization and significantly differentially expressed were selected, and the differential expression of genes was verified by real-time fluorescent quantitative reverse transcription polymerase chain reaction (RT-PCR). Data were statistically analyzed with independent-samples t test. Results: (1) The cultured cells were cloned with a paving stone-like shape and positive rate of CD49f of 95.132% and CD71 of 0.006%, which proved that the primary culture of ESCs was successful. (2) The quality control data analysis showed that the selected samples had better quality and higher sequence alignment rate, which met the requirements of sequencing. (3) Sequencing data showed that there were a total of 449 differentially expressed genes between the two groups, including 354 up-regulated genes and 95 down-regulated genes. Further cluster analysis determined that there were 18 significantly up-regulated genes and 5 significantly down-regulated genes between the two groups. GO enrichment analysis and KEGG pathway annotation analysis showed that the significantly differentially expressed genes were mainly enriched in the epidermis construction and keratinization process, which also might be related to interleukin 17 signaling pathway. (4) Keratin 19, keratin 10, and S100A7 genes which were related to the process of re-epithelialization and significantly differentially expressed were selected for verification. Real-time fluorescent quantitative RT-PCR showed that compared with those of control group, the mRNA expressions of keratin 19 and S100A7 of cells in PRP group were significantly increased (t=10.270, 5.690, P<0.01), while the mRNA expression of keratin 10 was significantly decreased (t=7.306, P<0.01), which was consistent with the result of sequencing data. Conclusions: PRP regulates function of human ESCs and promotes wound re-epithelialization involving transcriptional regulation of multiple genes, including keratin 19, keratin 10, and S100A7. In-depth exploration of the possible regulatory network of PRP affecting human ESCs will provide the basis for its subsequent clinical application.


Assuntos
Plasma Rico em Plaquetas , Reepitelização , Adolescente , Adulto , Criança , Pré-Escolar , Células Epidérmicas , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Células-Tronco , Transcriptoma , Adulto Jovem
11.
Zhonghua Shao Shang Za Zhi ; 36(10): 905-914, 2020 Oct 20.
Artigo em Chinês | MEDLINE | ID: mdl-33105942

RESUMO

Objective: To explore the mechanism of dendritic epidermal T lymphocytes (DETCs) in promoting healing of full-thickness skin defect wound on mice by regulating the proliferation and differentiation of epidermal stem cells (ESCs) in mice. Methods: (1) Ten 8-week-old wild type (WT) male C57BL/6 mice (the same sex and kind below) were sacrificed to collect the skin of back for extracting DETCs to culture. Five WT and five 8-week-old T cell receptor (TCR) δ(-)/(-) mice were selected and enrolled in WT control group and TCR δ(-)/(-) control group, respectively. A full-thickness skin defect wound with diameter of 6 mm was made on both sides of spinal line on the back of mice without any treatment after injury. Another fifteen 8-week-old TCR δ(-)/(-) mice were selected and divided into phosphate buffer solution (PBS), DETC, and insulin-like growth factor-Ⅰ(IGF-Ⅰ) groups according to the random number table (the same grouping method below), with 5 mice in each group, and the same full-thickness skin defect wound was made on each mouse. Immediately after injury, mice in PBS, DETC, and IGF-Ⅰ groups were injected subcutaneously around each wound with 10 µL sterile PBS , DETCs (cell concentration of 1×10(6)/mL), and 5 mg/mL recombinant mice IGF-Ⅰ, respectively. The percentage of the residual wound area was calculated on post injury day (PID) 2, 4, 6, and 8. (2) Three 8-week-old WT mice were enrolled in WT control group and nine 8-week-old TCR δ(-)/(-) mice were divided into TCR δ(-)/(-) control group, PBS group, and DETC group, with 3 mice in each group. The full-thickness skin defect wound was made as in experiment (1) . On PID 3, the protein expression of IGF-Ⅰ in the epidermis tissue of wound margin was detected by chemiluminescence imaging analyzer. (3) Three 8-week-old WT mice were enrolled in WT control group and six 8-week-old TCR δ(-)/(-) mice were divided into PBS and DETC groups, with 3 mice in each group, and the full-thickness skin defect wound was made as in experiment (1). On PID3, DETCs were extracted from the wound margin epidermis tissue to detect the percentage of DETCs expressing IGF-Ⅰ by flow cytometer. (4) The mice were taken as in experiment (2) and divided into WT control, PBS, DETC, and IGF-Ⅰ groups. A straight full-thickness skin defect incision with length of 3 cm was made in the direction of one inner ear. Mice in WT control group didn't have any other treatment after injury, and immediately after injury, mice in PBS, DETC, and IGF-Ⅰ groups were injected subcutaneously around each wound with 10 µL sterile PBS, DETCs (cell concentration of 1×10(6)/mL), and 5 mg/mL recombinant mice IGF-Ⅰ, respectively. On PID 12, epidermis tissue of wound margin was collected, and immunofluorescence staining was performed to observe the number of keratin 15 positive cells. (5) The same mice were collected, grouped, and treated as in experiment (4). On PID12, the epidermis tissue of wound margin was collected and immunofluorescence staining was performed to observe the number of keratin 10 positive cells. (6) Twenty 3-day-old WT mice (the same below) were sacrificed to collect the whole skin, which was used to extract ESCs, with 5 mice detecting one index. The ESCs were divided into DETC co-culture group and control group, which were added with 1 mL DETCs (cell concentration of 1.25×10(6)/mL) and DETC medium, respectively. The percentage of 5-ethynyl-2'-deoxyuridine (EdU) positive cell on culture day (CD) 3, the percentages of CD49f(+) CD71(-) and keratin 14 positive cells on CD 5, and the percentage of keratin 10 positive cell on CD 10 in 2 groups were detected by flow cytometer. (7) Twenty mice were taken to extract ESCs, with 5 mice detecting one index. The ESCs were divided into control group and IGF-Ⅰ group, which were added with 1 mL sterile PBS and 10 ng/mL recombinant mice IGF-Ⅰ, respectively. The percentages of EdU positive cell, CD49f(+) CD71(-) cell, keratin10 positive cell, and keratin 14 positive cell were detected as in experiment (6). The sample in each group of experiments (6) and (7) was three. Data were statistically analyzed with analysis of variance for repeated measurement, one-way analysis of variance, and t test. Results: (1) On PID 4, 6, and 8, the percentage of residual wound area in TCR δ(-)/(-) control group was significantly higher than that in WT control group (t=2.78, 3.39, 3.66, P<0.05 or P<0.01). The percentage of residual wound area in DETC group and IGF-Ⅰgroup on PID 4, 6, and 8 was apparently lower than that in PBS group (t=2.61, 3.21, 3.88, 2.84, 2.91, 2.49, P<0.05 or P<0.01). (2) On PID 3, the protein expression of IGF-Ⅰ in the epidermis tissue of wound margin of mice in TCR δ(-)/(-) control group was significantly lower than that in WT control group (t=17.34, P<0.01). The protein expression of IGF-Ⅰ in the epidermis tissue of wound margin of mice in DETC group was significantly higher than that in PBS group (t=11.71, P<0.01). (3) On PID 3, the percentage of DETCs expressing IGF-Ⅰ in the epidermis tissue of wound margin of mice in PBS group was significantly lower than that in WT control group and DETC group (t=24.95, 27.23, P<0.01). (4) On PID 12, the number of keratin 15 positive cells in the epidermis tissue of wound margin of mice in PBS group was significantly lower than that in WT control group, DETC group, and IGF-Ⅰ group (t=17.97, 11.95, 7.63, P<0.01). (5) The number of keratin 10 positive cells in the epidermis tissue of wound margin of mice in PBS group was significantly higher than that in WT control group, DETC group, and IGF-Ⅰ group (t=11.59, 9.51, 3.48, P<0.05 or P<0.01). (6) The percentages of EdU positive cells on CD 3, CD49f(+) CD71(-) cells on CD 5, and keratin 14 positive cells on CD 5 in DETC co-culture group were respectively (43.5±0.6)%, (66.5±0.5)%, (69.3±1.7)%, apparently higher than (32.3±1.3)%, (56.4±0.3)%, (54.9±1.3)% in control group (t=7.97, 17.10, 6.66, P<0.01). The percentage of keratin 10 positive cells on CD 10 in DETC co-culture group was (55.7±0.7)%, significantly lower than (67.1±1.2)% in control group (t=8.34, P<0.01). (7) The percentages of EdU positive cells on CD 3, CD49f(+) CD71(-) cells on CD 5, and keratin 14 positive cells on CD 5 in IGF-Ⅰ group were respectively (42.1±0.9)%, (81.1±1.3)%, (66.8±1.0)%, apparently higher than (32.4±0.7)%, (74.9±0.7)%, (52.0±1.9)% in control group (t=8.39, 4.24, 7.25, P<0.05 or P<0.01). The percentage of keratin 10 positive cells on CD 10 in IGF-Ⅰ group was (53.5±1.1)% , significantly lower than (58.2±0.3)% in control group (t=3.99, P<0.05). Conclusions: DETCs can promote the proliferation and anti-apoptotic potential of ESCs and inhibit their differentiation into end-stage by secreting IGF-Ⅰ, thus promoting wound healing of full-thickness skin defects in mice.


Assuntos
Linfócitos T , Cicatrização , Animais , Diferenciação Celular , Proliferação de Células , Epiderme , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco
12.
Zhonghua Shao Shang Za Zhi ; 36(10): 923-929, 2020 Oct 20.
Artigo em Chinês | MEDLINE | ID: mdl-33105944

RESUMO

Objective: To investigate the mechanisms of interleukin-17A (IL-17A) regulating the expressions of IL-1ß and IL-23 in mouse keratinocytes (KCs). Methods: Primary KCs were isolated from the skin of 400 newborn male and female wild type C57BL/6 mice and cultured in 24-well plates with Roswell Park Memorial Institute 1640 medium containing fetal bovine serum in the volume fraction of 10% for the following experiments. (1) The cells were divided into phosphate buffer solution (PBS) control group and IL-17A stimulation group according to the random number table (the same grouping method below), which were cultured with 10 µL PBS or 10 µL IL-17A in the mass concentration of 100 ng/mL for 6 hours, respectively. The expression levels of IL-1ß and IL-23 mRNA in cells were detected by real-time fluorescence quantitative reverse transcription polymerase chain reaction (RT-PCR), with 3 samples in each group. (2) The cells were divided into dimethyl sulfoxide (DMSO) control group, IL-17A+ DMSO group, IL-17A+ nuclear factor κB (NF-κB) inhibitor group, IL-17A+ signal transduction and activator of transcription 3 (STAT3) inhibitor group, IL-17A+ extracellular signal-regulated kinase 1 (ERK1) inhibitor group, IL-17A+ ERK2 inhibitor group, and IL-17A+ c-Jun N-terminal kinase (JNK) inhibitor group. The reagents were added to cells in corresponding groups respectively and cultured for 6 hours. The volume of each reagent was 10 µL, the mass concentration of IL-17A was 100 ng/mL, and the molarity concentrations of NF-κB, STAT3, ERK1, ERK2, JNK signal pathway inhibitors PDTC, S3I-201, SCH772984, SCH772984, SP600125 were 5 µmol/L, 100 µmol/L, 4 nmol/L, 1 nmol/L, and 10 µmol/L, respectively. The expression levels of IL-1ß mRNA and IL-23 mRNA in cells were detected by real-time fluorescence quantitative RT-PCR, with 3 samples in each group. (3) The cells were grouped and treated the same as those in experiment (1). The levels of NF-κB phosphorylation, STAT3 phosphorylation, ERK phosphorylation, and JNK phosphorylation were detected by Western blotting, with 3 samples in each group. Data were statistically analyzed with two-tailed Student t test, one-way analysis of variance, t test, and Bonferroni correction. Results: (1) After culture of 6 hours, compared with those in PBS control group, the expression levels of IL-1ß and IL-23 mRNA in cells in IL-17A stimulation group were significantly increased (t=13.46, 6.72, P<0.01). (2) After culture of 6 hours, the expression levels of IL-1ß and IL-23 mRNA in cells in DMSO control group, IL-17A+ DMSO group, IL-17A+ NF-κB inhibitor group, IL-17A+ STAT3 inhibitor group, IL-17A+ ERK1 inhibitor group, IL-17A+ ERK2 inhibitor group, and IL-17A+ JNK inhibitor group were 1.00±0.11, 4.01±0.32, 0.32±0.06, 1.76±0.43, 3.62±0.24, 3.80±0.43, 4.26±0.74 and 1.03±0.29, 4.08±0.34, 4.76±0.38, 4.70±0.21, 1.06±0.42, 0.92±0.21, 0.39±0.05, respectively. Compared with those in DMSO control group, the expression levels of IL-1ß and IL-23 mRNA in cells in IL-17A+ DMSO group were significantly increased (t=9.24, 12.60, P<0.01). Compared with that in IL-17A+ DMSO group, the expression level of IL-1ß mRNA was significantly decreased in cells in IL-17A+ NF-κB inhibitor group and IL-17A+ STAT3 inhibitor group (t=11.34, 6.91, P<0.01). Compared with that in IL-17A+ DMSO group, the expression level of IL-23 mRNA was significantly decreased in cells in IL-17A+ ERK1 inhibitor group, IL-17A+ ERK2 inhibitor group, and IL-17A+ JNK inhibitor group (t=12.44, 13.03, 15.21, P<0.01). (3) After culture of 6 hours, compared with those in PBS control group, the levels of NF-κB phosphorylation, STAT3 phosphorylation, ERK phosphorylation, and JNK phosphorylation in cells in IL-17A stimulation group were significantly increased. Conclusions: IL-17A promotes the transcription of IL-1ß in mouse KCs through the phosphorylation of NF-κB and STAT3 pathways and IL-23 through the phosphorylation of ERK and JNK pathways.


Assuntos
Interleucina-17 , Interleucina-23 , Animais , Interleucina-1beta , Queratinócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B
13.
Zhonghua Shao Shang Za Zhi ; 36(3): 224-233, 2020 Mar 20.
Artigo em Chinês | MEDLINE | ID: mdl-32241049

RESUMO

Objective: To explore the transcriptional regulation mechanism of transforming growth factor ß(1) (TGF-ß(1)) on Meox1 and its effect on cell migration of adult human dermal fibroblasts (HDF-a). Methods: (1) HDF-a cells were cultured in RPMI 1640 complete medium (hereinafter referred to as routinely cultured). The cells were divided into TGF-ß(1) stimulation group and blank control group. The cells in TGF-ß(1) stimulation group were stimulated with 10 µL TGF-ß(1) in the mass concentration of 1 mg/µL, while the cells in blank control group were stimulated with the equal volume of phosphate buffer solution. After 72 hours in culture, partial cells in both groups were collected for transcriptome sequencing. The genes with differential expression ratio greater than or equal to 2 and P<0.01 between the two groups were selected to perform enrichment analysis and analysis of metabolic pathways of the Kyoto Gene and Genome Encyclopedia with, and the expression value of Meox1 per million transcripts (TPM) was recorded (n=3). Partial cells from the two groups were used to detect the Meox1 mRNA expression by real-time fluorescent quantitative reverse transcription polymerase chain reaction (RT-PCR) (n=3). (2) Cultured HDF-a cells in the logarithmic growth phase (the same growth phase of cells below) were divided into empty plasmid group, Smad2 overexpression (OE) group, Smad3 OE group, and Smad4 OE group, which were transfected respectively with 2 µg empty pcDNA3.1 plasmid and pcDNA3.1 plasmids separately carrying Smad2, Smad3, and Smad4 for 6 hours, and then were routinely cultured for 48 hours. The Meox1 mRNA expression in the transfected cells of each group was detected by real-time fluorescent quantitative RT-PCR (n=3). (3) HDF-a cells were routinely cultured and grouped the same as in experiment (1). After 72 hours in culture, the enrichment of Smad2, Smad3, and Smad4 protein on the Meox1 promoter in the cells of each group was detected by chromatin immunoprecipitation-quantitative PCR (ChIP-qPCR) (n=3). (4) HDF-a cells were routinely cultured and divided into negative interference group, small interference RNA (siRNA)-Smad2 group, siRNA-Smad3 group, siRNA-Smad4 group, empty plasmid group, Smad2 OE group, Smad3 OE group, and Smad4 OE group, which were transfected respectively with 50 µmol/L random siRNA, siRNA-Smad2, siRNA-Smad3, siRNA-Smad4, 2 µg empty pcDNA3.1 plasmid and pcDNA3.1 plasmids separately carrying Smad2, Smad3, and Smad4 for 6 hours and then routinely cultured for 48 hours. The enrichment of Smad2, Smad3, and Smad4 protein on the Meox1 promoter in the cells of corresponding group was detected by ChIP-qPCR (n=3). (5) Two batches of HDF-a cells were cultured and divided into negative interference group, siRNA-Meox1 group, empty plasmid group, and Meox1 OE group, which were transfected respectively with 50 µmol/L random siRNA, siRNA-Meox1, 2 µg empty pcDNA3.1 plasmid and pcDNA3.1 plasmid carrying Meox1 for 6 hours and then routinely cultured for 24 hours. One batch of cells were subjected to scratch test with the scratch width being observed 24 hours after scratching and compared with the initial width for scratch wound healing; the other batch of cells were subjected to Transwell assay, in which the migrated cells were counted after being routinely cultured for 24 hours (n=3). (6) From January 2018 to June 2019, 3 hypertrophic scar patients (2 males and 1 female, aged 35-56 years) were admitted to the First Affiliated Hospital of Army Medical University (the Third Military Medical University) 8-12 months after burns. The scar tissue and normal skin tissue along the scar margin resected during surgery were taken, and immunohistochemical staining was performed to observe the distribution of Meox1 protein expression. Data were statistically analyzed with one-way analysis of variance and independent sample t test. Results: (1) After 72 hours in culture, a total of 843 genes were obviously differentially expressed between the two groups, being related to tissue repair, cell migration, inflammatory cell chemotaxis induction process and potential signaling pathways such as tumor necrosis factor, interleukin 17, extracellular matrix receptor. The TPM value of Meox1 in the cells of blank control group was 45.9±1.9, which was significantly lower than 163.1±29.5 of TGF-ß(1) stimulation group (t=6.88, P<0.01) with RNA-sequencing. After 72 hours in culture, the Meox1 mRNA expression levels in the cells of blank control group was 1.00±0.21, which was significantly lower than 11.00±3.61 of TGF-ß(1) stimulation group (t=4.79, P<0.01). (2) After 48 hours in culture, the Meox1 mRNA expression levels in the cells of Smad2 OE group, Smad3 OE group, and Smad4 OE group were 198.70±11.02, 35.47±4.30, 20.27±2.50, respectively, which were significantly higher than 1.03±0.19 of empty plasmid group (t=31.07, 13.80, 13.12, P<0.01). (3) After 72 hours in culture, the enrichment of Smad2, Smad3, and Smad4 protein on the promoter of Meox1 in the cells of TGF-ß(1) stimulation group was significantly higher than that of blank control group respectively (t=12.99, 41.47, 29.10, P<0.01). (4) After 48 hours in culture, the enrichment of Smad2 protein on the promoter of Meox1 in the cells of negative interference group was (0.200 000±0.030 000)%, significantly higher than (0.000 770±0.000 013)% of siRNA-Smad2 group (t=11.67, P<0.01); the enrichment of Smad2 protein on the promoter of Meox1 in the cells of empty plasmid group was (0.200 000±0.040 000)%, significantly lower than (0.700 000±0.090 000)% of Smad2 OE group (t=8.85, P<0.01). The enrichment of Smad3 protein on the promoter of Meox1 in the cells of negative interference group was (0.500 0±0.041 3)%, significantly higher than (0.006 0±0.001 3)% of siRNA-Smad3 group (t=17.79, P<0.01); the enrichment of Smad3 protein on the promoter of Meox1 in the cells of empty plasmid group was (0.470 0±0.080 0)%, which was significantly lower than (1.100 0±0.070 0)% of Smad3 OE group (t=9.93, P<0.01). The enrichment of Smad4 protein on the promoter of Meox1 in the cells of negative interference group was similar to that of siRNA-Smad4 group (t=2.11, P>0.05); the enrichment of Smad4 protein on the promoter of Meox1 in the cells of empty plasmid group was similar to that of Smad4 OE group (t=0.60, P>0.05). (5) Twenty-four hours after scratching, the scratch healing width of cells in siRNA-Meox1 group was narrower than that of negative interference group, while that of Meox1 OE group was wider than that of empty plasmid group. After 24 hours in culture, the number of migration cells in negative interference group was significantly higher than that in siRNA-Meox1 group (t=9.12, P<0.01), and that in empty plasmid group was significantly lower than that in Meox1 OE group (t=8.99, P<0.01). (6) The expression of Meox1 protein in the scar tissue was significantly higher than that in normal skin of patients with hypertrophic scars. Conclusions: TGF-ß(1) transcriptionally regulates Meox1 expression via Smad2/3 in HDF-a cells, thus promoting cell migration.


Assuntos
Movimento Celular , Cicatriz Hipertrófica , Fibroblastos/metabolismo , Proteínas de Homeodomínio , Fatores de Transcrição , Fator de Crescimento Transformador beta1/metabolismo , Adulto , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Transdução de Sinais , Fator de Crescimento Transformador beta
14.
Zhonghua Shao Shang Za Zhi ; 36(2): 122-130, 2020 Feb 20.
Artigo em Chinês | MEDLINE | ID: mdl-32114730

RESUMO

Objective: To explore the effects of dendritic epidermal T cells (DETC) on proliferation and apoptosis of epidermal cells in wound margin of mice and its effects on wound healing. Methods: Twenty-eight healthy specific pathogen free (SPF) C57BL/6 wild-type (WT) male mice aged 8-12 weeks and 60 SPF T lymphocyte receptor δ-knockout (TCR δ(-/-)) male mice aged 8-12 weeks were selected to conduct the following experiments. (1) Eight WT mice were selected to isolate epidermal cells and primarily culture DETC according to the random number table. Morphological observation and purity identification of DETC by flow cytometer were detected immediately after culture and on culture day (CD) 15 and 30, respectively. (2) According to the random number table, 5 WT mice and 5 TCR δ(-/-) mice were selected and enrolled into WT control group and TCR δ(-/-) group. Round full-thickness skin defect with diameter of 6 mm was made on the back of each mouse. The wound healing condition was observed immediately after injury and on post injury day (PID) 2, 4, 6, 8, 10, and the percentage of residual wound area was calculated. (3) Mice were selected to group and reproduce model of full-thickness skin defect as in experiment (2). On PID 3, the tissue of wound margin was collected for hematoxylin eosin staining, and the length of new epithelium was measured. (4) Mice were selected to group and reproduce model of full-thickness skin defect as in experiment (2). On PID 3, epidermal tissue of wound margin was collected to determine expression of proliferating cell nuclear antigen (PCNA) using Western blotting for evaluation of proliferation of epidermal cell. (5) Mice were selected to group and reproduce model of full-thickness skin defect as in experiment (2). On PID 3, epidermal tissue of wound margin was selected and digested into single-cell suspension, and apoptosis of cells was detected by flow cytometer. (6) Forty TCR δ(-/-) mice were selected to carry out the same treatment as in experiments (2)-(5). According to the random number table, these mice were enrolled into TCR δ(-/-) control group and TCR δ(-/-)+ DETC group, with 5 mice in each group for each experiment. Round full-thickness skin defect was made on the back of each mouse. DETC in the number of 1×10(5) (dissolution in 100 µL phosphate with buffer purity above 90%) were injected through multiple points of wound margin of mice in TCR δ(-/-)+ DETC group immediately after injury, and equal volume of phosphate buffer was injected into mice of TCR δ(-/-) control group with the same method as above. Data were processed with one-way analysis of variance for repeated measurement, t test, and Bonferroni correction. Results: (1) Along with the culture time elapse, the number of dendritic structures of DETC increased gradually. The percentage of T lymphocytes was 4.67% and 94.1% of these T lymphocytes were DETC. The purity of DETC on CD 15 was 18.50% and the purity of DETC on CD 30 was 98.70%. (2) Immediately after injury, the wound healing condition of mice in WT control group and TCR δ(-/-) group was similar. The wound healing speed of mice in TCR δ(-/-) group was slower than that in WT control group on PID 2-10. The percentages of residual wound area of mice in TCR δ(-/-) group on PID 2, 4, 6, 8, and 10 were increased significantly compared with those in WT control group (t=3.492, 4.425, 4.170, 4.780, 7.318, P<0.01). (3) The length of new epithelium of mice in TCR δ(-/-) group on PID 3 was (359 ± 15) µm, which was obviously shorter than that in WT control group [(462±26) µm, t=3.462, P<0.01]. (4) Immediately after injury, wound condition of mice in TCR δ(-/-)+ DETC group and TCR δ(-/-) control group was similar. Compared with TCR δ(-/-)+ DETC group, the wound healing speed of mice in TCR δ(-/-) control group were obviously slower on PID 2-10. The percentages of residual wound area of mice in TCR δ(-/-)+ DETC group on PID 2, 4, 6, 8, and 10 were decreased significantly compared with those in TCR δ(-/-) control group (t=2.308, 3.725, 2.698, 3.707, 6.093, P<0.05 or P<0.01). (5) On PID 3, the length of new epithelium of mice in TCR δ(-/-)+ DETC group was (465±31) µm, which was obviously longer than that in TCR δ(-/-) control group [(375±21) µm, t=2.390, P<0.05]. (6) On PID 3, PCNA expression of epidermal cell in wound margin of mice in TCR δ(-/-) group was 1.25±0.04, which was obviously lower than that in WT control group (2.01±0.09, t=7.415, P<0.01). (7) On PID 3, PCNA expression of epidermal cell in wound margin of mice in TCR δ(-/-)+ DETC group was 1.62±0.08, which was significantly higher than that in TCR δ(-/-) control group (1.05±0.14, t=3.561, P<0.05). (8) On PID 3, apoptosis rate of epidermal cell in wound margin of mice in TCR δ(-/-) group was (16.1±1.4)%, which was higher than that in WT control group [(8.1±0.6)%, t=5.363, P<0.01]. (9) On PID 3, apoptosis rate of epidermal cell in wound margin of mice in TCR δ(-/-)+ DETC group was (11.4±1.0)%, which was obviously lower than that in TCR δ(-/-) control group [(15.4±1.4)%, t=2.377, P<0.05]. Conclusions: DETC participates in the process of wound healing though promoting the proliferation of epidermal cells in wound margin and inhibit the apoptosis of these cells.


Assuntos
Células Epidérmicas , Linfócitos T , Animais , Apoptose , Proliferação de Células , Células Dendríticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
Zhonghua Shao Shang Za Zhi ; 36(6): 471-475, 2020 Jun 20.
Artigo em Chinês | MEDLINE | ID: mdl-32114747

RESUMO

Coronavirus disease 2019 (COVID-19) has seriously affected the safety of patients and social stability. Some COVID-19 patients in the later stage of disease may develop into acute respiratory distress syndrome or even multiple organ failure. However, one of the most important mechanisms underlying the deterioration of disease is cytokine storm. At present, some therapies such as interleukin-6 antibody blocker, stem cell therapy, and transfusion of convalescent plasma have been applied to counteract the cytokine storm with some progresses being achieved. This article reviews the influences of cytokine storm syndrome on the COVID-19 and the corresponding immunotherapies to resist cytokine storm.


Assuntos
Infecções por Coronavirus/imunologia , Infecções por Coronavirus/terapia , Síndrome da Liberação de Citocina/terapia , Imunoterapia , Pneumonia Viral/imunologia , Pneumonia Viral/terapia , Anticorpos/uso terapêutico , Betacoronavirus , COVID-19 , Síndrome da Liberação de Citocina/virologia , Humanos , Imunização Passiva , Interleucina-6/antagonistas & inibidores , Pandemias , SARS-CoV-2 , Transplante de Células-Tronco , Soroterapia para COVID-19
16.
Zhonghua Shao Shang Za Zhi ; 36(8): 691-697, 2020 Aug 20.
Artigo em Chinês | MEDLINE | ID: mdl-32174095

RESUMO

Coronavirus disease 2019 (COVID-19) outbroke in Wuhan, China in December 2019 and the severe acute respiratory syndrome (SARS) outbroke in Guangzhou, China in 2003 were caused by highly pathogenic coronaviruses with high homology. Since the 2019 novel coronavirus is highly contagious and spreads rapidly. It has caused negative social effects and massive economic loss globaly. Currently there is no vaccine or effective drugs. Pulmonary fibrosis is a pulmonary disease with progressive fibrosis, which is the main factor leading to pulmonary dysfunction and declined quality of life in SARS survivors after recovery. Extensive epidemiological, viral immunological and current clinical evidences support the possibility that pulmonary fibrosis may be one of the major complications in COVID-19 patients. At present there is no report on the mechanism by which COVID-19 induces pulmonary fibrosis.With the existing theoretical basis, this article focuses on discussing the possible mechanism of COVID-19 sustained lung damaging, the key role of abnormal immune mechanism in the initiation and promotion of pulmonary fibrosis, and the corresponding therapeutic measures.


Assuntos
Betacoronavirus , Infecções por Coronavirus , Pandemias , Pneumonia Viral , Fibrose Pulmonar , COVID-19 , China , Infecções por Coronavirus/complicações , Humanos , Pneumonia Viral/complicações , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/terapia , Qualidade de Vida , SARS-CoV-2
17.
Eur Rev Med Pharmacol Sci ; 23(23): 10541-10548, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31841210

RESUMO

OBJECTIVE: To explore the influence of butorphanol on myocardial ischemia/reperfusion (I/R) injury in rats through the mitogen-activated protein kinase (MAPK) signaling pathway. MATERIALS AND METHODS: The I/R model in Sprague-Dawley rats was established. The rats were randomly divided into normal group (n=20), myocardial I/R model group (model group, n=20), and butorphanol treatment group (treatment group, n=20). Next, the liver function indicators such as alkaline phosphatase (ALP), alanine aminotransferase (ALT), and the myocardial function index creatine kinase (CK) in rats were detected. ELISA was carried out to measure the relative levels of tumor necrosis factor-gamma (TNF-γ), interleukin-6 (IL-6), and IL-1α in serum samples of rats. The cardiac function indexes were examined via magnetic resonance imaging (MRI) and echocardiography (ECG). Besides, the pathological changes of the myocardial tissues were detected through hematoxylin-eosin (HE) staining. The quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) and Western blotting were performed to measure the mRNA and protein expression levels of the relative genes in the MAPK signaling pathway in the rat myocardial tissues. RESULTS: The serum levels of ALP, ALT, and CK in I/R model group were significantly higher than those in the normal group. In I/R model group, the relative levels of TNF-γ, IL-6, and IL-1α, as well as left ventricular end-diastolic diameter (LVEDd) and left ventricular end-systolic diameter (LVESd), were remarkably higher, while the fractional shortening (FS, %) and the ejection fraction (EF, %) were lower in comparison with those in the normal group. The HE staining results showed that the myocardial tissues in the I/R model group exhibited severe injury. The expression levels of Caspase3, MAPK, and c-Jun N-terminal kinase (JNK) were clearly higher in the I/R model group than those in the treatment group (p<0.05), while the expression level of extracellular regulated protein kinase 1 (ERK1) was remarkably lower (p<0.05). The protein level of MAPK in the treatment group was overtly reduced compared with that in the I/R model group (p<0.05). CONCLUSIONS: Butorphanol can modulate the recovery of the myocardial injury in the rats after the myocardial I/R by inhibiting the MAPK signaling pathway.


Assuntos
Butorfanol/administração & dosagem , Cardiotônicos/administração & dosagem , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Humanos , Masculino , Infarto do Miocárdio/complicações , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/diagnóstico , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Volume Sistólico/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos
18.
Zhonghua Shao Shang Za Zhi ; 35(7): 557-559, 2019 Jul 20.
Artigo em Chinês | MEDLINE | ID: mdl-31357831

RESUMO

The 15th Syposium on Chinese Burn Medicine and the 2nd Congress of Burn Medicine Branch of China International Exchange and Promotion Association for Medical and Healthcare (CPAM) was successfully held in Suzhou, from June 20th to 22th in 2019. A total of 400 specialists and scholars across the country attended the meeting. Focusing on the theme of " Guide and consensus: exploration and consideration " , with form of one main meeting place and two branch meeting places, the related hot and difficult problems were discussed warmly. During the conference, Working Conference of Editorial Committee of Chinese Journal of Burns, Standing Committee of the Chinese Burn Association, and the Congress of Burn Medicine Branch of CPAM were held.


Assuntos
Queimaduras , China , Congressos como Assunto , Humanos
19.
Zhonghua Shao Shang Za Zhi ; 35(4): 298-307, 2019 Apr 20.
Artigo em Chinês | MEDLINE | ID: mdl-31060178

RESUMO

Objective: To explore effects of dendritic epidermal T cells (DETCs) and Vγ4 T lymphocytes on proliferation and differentiation of mice epidermal cells and the effects in wound healing of mice. Methods: (1) Six C57BL/6 male mice aged 8 weeks were collected and divided into control group and wound group according to random number table (the same grouping method below), with 3 mice in each group. A 4 cm long straight excision with full-thickness skin defect was cut on back of each mouse in wound group, while mice in control group received no treatment. On post injury day (PID) 3, mice in 2 groups were sacrificed, and skin within 5 mm from the wound margin on back of mice in wound group and normal skin on corresponding part of mice in control group were collected to make single cell suspensions. The percentage of Vγ4 T lymphocyte expressing interleukin-17A (IL-17A) and percentage of DETCs expressing insulin-like growth factor Ⅰ (IGF-Ⅰ) were detected by flow cytometer. (2) Ten C57BL/6 male mice aged 8 weeks were collected and divided into control group and Vγ4 T lymphocyte depletion group with 5 mice in each group. Mice in Vγ4 T lymphocyte depletion group were injected with 200 g Vγ4 T lymphocyte monoclonal neutralizing antibody of Armenian hamster anti-mouse intraperitoneally, and mice in control group were injected with the same amount of Armenian hamster Ig intraperitoneally. One hole with full-thickness skin defect was made on each side of spine of back of each mice. The wound healing was observed on PID 1-8, and percentage of remaining wound area was calculated. (3) Six C57BL/6 male mice aged 8 weeks were grouped and treated in the same way as in experiment (2), with 3 mice in each group. On PID 3, expressions of IL-17A and IGF-Ⅰ in epidermis on margin of wound were detected with Western blotting. (4) Thirty C57BL/6 male mice aged 3 days were sacrificed, and epidermal cells were extracted. The keratin 14 positive cell rate was examined by flow cytometer (the same detecting method below). (5) Another batch of mouse epidermal cells were collected and divided into control group, IGF-Ⅰ group, and IL-17A group, with 3 wells in each group (the same well number below). Cells in IGF-Ⅰ group and IL-17A group were added with 1 mL recombinant mouse IGF-Ⅰ and IL-17A with final mass concentration of 100 ng/mL respectively, while cells in control group were added with the same amount of sterile phosphate buffered saline (PBS). On post culture day (PCD) 5, keratin 14 negative cell rate was examined. Another batch of mouse epidermal cells were collected, grouped, and treated in the same way as aforementioned experiment, and keratin 10 positive cell rate was examined on PCD 10. (6) Another batch of mouse epidermal cells were collected and added with 4 mmol/L 5(6)-carboxyfluorescein diacetate N-succinimidyl ester (CFSE) solution, and divided into control 0 d group, control 7 d group, IGF-Ⅰ group, and IL-17A group. Cells in IGF-Ⅰ group and IL-17A group were treated in the same way as the corresponding groups in experiment (5), and cells in control 0 d group and control 7 d group were treated in the same way as the control group in experiment (5). The CFSE fluorescence peaks were examined on PCD 0 of control 0 d group and PCD 7 of the other 3 groups. (7) Another batch of mouse epidermal cells were collected and divided into control group and IGF-Ⅰ group. Cells in IGF-Ⅰ group were added with 1 mL recombinant mouse IGF-Ⅰ with final mass concentration of 100 ng/mL, and cells in control group were added with the same amount of sterile PBS. On PCD 5, cells were underwent keratin 14 staining and CFSE staining as aforementioned, and keratin 14 negative cell rate of CFSE positive cells was examined. Another batch of mouse epidermal cells were collected and divided into control group and IL-17A group. Cells in IL-17A group were added with 1 mL recombinant mouse IL-17A with final mass concentration of 100 ng/mL, and cells in control group were added with the same amount of sterile PBS. On PCD 5, keratin 14 negative cell rate of CFSE positive cells was examined. Data were processed with one-way analysis of variance and t test. Results: (1) On PID 3, percentage of DETC expressing IGF-Ⅰ in normal epidermis of control group was (9.9±0.8)%, significantly lower than (19.0±0.6)% of epidermis around margin of wound group (t=8.70, P<0.01); percentage of Vγ4 T lymphocyte expressing IL-17A in normal epidermis of control group was (0.123±0.024)%, significantly lower than (8.967±0.406)% of epidermis around margin of wound group (t=21.77, P<0.01). (2) On PID 1-4, there was obvious inflammatory reaction around wounds of mice in control group, and on PID 5-8, the wound area was still large. On PID 1-4, there was slight inflammatory reaction around wounds of mice in Vγ4 T lymphocyte depletion group, and on PID 5-8, the wound area was significantly reduced. On PID 3-7, percentages of residual wound area in Vγ4 T lymphocyte depletion group were significantly lower than those in control group (t=5.92, 5.74, 7.17, 5.38, 5.57, P<0.01), while percentages of residual wound area in two groups on PID 1, 2, 6 were similar (t=1.46, 3.17, 3.10, P>0.05). (3) On PID 3, compared with those in control group, expression of IL-17A and IGF-Ⅰ in epidermis around wound margin of mice in Vγ4 T lymphocyte depletion group was markedly decreased and increased respectively (t=8.47, 19.24, P<0.01). (4) The keratin 14 positive cell rate of mouse epidermal cells was 94.7%. (5) On PCD 5, the keratin 14 negative cell rate of mice in control group was markedly higher than that of IGF-Ⅰ group, while significantly lower than that of IL-17A group (t=7.25, 5.64, P<0.01). On PCD 10, the keratin 10 positive cell rate of mice in control group was significantly higher than that of IGF-Ⅰ group, while significantly lower than that of IL-17A group (t=3.99, 10.82, P<0.05 or P<0.01). (6) Compared with that of control 0 d group, CFSE fluorescence peaks of mouse epidermal cells in control 7 d group, IGF-Ⅰ group, and IL-17A group on PCD 7 shifted to the left. Compared with that of control 7 d group, CFSE fluorescence peaks of mouse epidermal cells in IGF-Ⅰ group and IL-17A group on PCD 7 shifted to the left. (7) On PCD 5, keratin 14 negative cell rate of CFSE positive cells of mice in control group was significantly higher than that in IGF-Ⅰ group (t=9.91, P<0.01), and keratin 14 negative cell rate of CFSE positive cells of mice in control group was markedly lower than that in IL-17A group (t=6.49, P<0.01). Conclusions: In the process of wound healing, IGF-Ⅰ secreted by DETC can promote the proliferation of mouse keratin 14 positive epidermal cells and inhibit their terminal differentiation, while IL-17A secreted by Vγ4 T lymphocyte can promote the proliferation and terminal differentiation of mouse keratin 14 positive epidermal cells, thus both IGF-Ⅰ and IL-17A can affect wound healing.


Assuntos
Células Epidérmicas , Linfócitos T/imunologia , Cicatrização/imunologia , Animais , Diferenciação Celular , Proliferação de Células , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/metabolismo
20.
Zhonghua Shao Shang Za Zhi ; 33(5): 287-294, 2017 May 20.
Artigo em Chinês | MEDLINE | ID: mdl-28651420

RESUMO

Objective: To explore the effects of hypoxia inducible factor-1α (HIF-1α) on P311 and its influence on the migration of murine epidermal stem cells (ESCs) under hypoxia in vitro. Methods: Two kinds of murine ESCs were isolated and obtained from 15 neonatal wild-type C57BL/6J mice and 5 congeneric source P311 gene knock-out mice, respectively. The first passage of cells were used in the following experiments after morphologic observation and detection of expression of cell surface markers CD71 and CD49f with flow cytometer. (1) After cell scratch assay, according to the random number table (the same dividing method below), ESCs of P311 gene knock-out mice were divided into normoxia group (cells were cultured with complete medium in normoxic carbon dioxide incubator, and the subsequent normoxic treatments were the same) and hypoxia group (cells were cultured in hypoxic carbon dioxide incubator containing 1% oxygen, and the subsequent hypoxic treatments were the same), with 12 inserts in each group. ESCs of wild-type mice were divided into normoxia group, pure hypoxia group, dimethyl sulfoxide (DMSO) control group (2 µL DMSO solvent was added for 1 h of normoxia treatment before hypoxia treatment), HIF-1α inhibitor group (cells were treated with 11 µmol/L HIF-1 inhibitor of 2 µL under normoxia condition for 1 h before hypoxia treatment), HIF-1α stabilizer group (the cells were treated with 2 µmol/L FG-4592 of 2 µL under normoxia condition for 1 h before hypoxia treatment), with 12 inserts in each group. Three inserts of each time point in each group were adopted respectively to measure the residual width of scratch under inverted phase contrast microscope at post scratch hour (PSH) 0 (immediately), 12, 24, and 48. (2) After hypoxia treatment, the protein level of HIF-1α in ESCs of wild-type mice was detected by Western blotting at post hypoxia hour (PHH) 0, 12, 24, and 48. (3) ESCs of wild-type mice were divided into pure hypoxia group, DMSO control group, HIF-1α inhibitor group, and HIF-1α stabilizer group as that of experiment (1) with the same treatment. The mRNA expression of P311 and expression of P311 in ESCs were determined by real-time fluorescent quantitative reverse transcription polymerase chain reaction and immunocytochemical staining, respectively, at PHH 0 (immediately), 12, 24, and 48 (with sample numbers of 12). (4) The second passage of human embryonic kidney 293 (HEK-293) cells were divided into empty vector hypoxia group (cells were cultured under hypoxia condition after being transfected with empty vector plasmid), P311 normoxia group (cells were cultured under normoxia condition after being transfected with P311 reporter gene plasmid), P311 hypoxia group (cells were cultured under hypoxia condition after being transfected with P311 reporter gene plasmid), P311 hypoxia+ HIF-1α inhibitor group (cells which were incubated with HIF-1α inhibitor were cultured under hypoxia condition after being transfected with P311 reporter gene plasmid). The luciferase activity was detected at post culture hour (PCH) 0 and 12, respectively, and then the P311 transcriptional regulatory binding site of HIF-1α and the promoter sequence of P311 were predicted and searched by bioinformatics methods. Data were processed with factorial design variance analysis, one-way analysis of variance, LSD test and Bonferroni correction. Results: (1) The results of ESCs. The cells showed cobblestone-like pattern and different clonal morphology due to the different cell proliferation potential. The proportion of CD71(-)CD49f(+) cells accounted for about 85%. The identification results indicated that the cells showed strong stem cell properties and high purity. Compared with those in cells of normoxia group of P311 gene knock-out mice, the residual widths of scratch of cells in pure hypoxia group were smaller at PSH 12 and 24 (with P values below 0.05), and those in hypoxia group, normoxia group of wild-type mice, DMSO control group, HIF-1α inhibitor group, and HIF-1α stabilizer group were smaller at PSH 12 (with P values below 0.05). Compared with those in cells of normoxia group of wild-type mice, the residual widths of scratch of cells in hypoxia group, pure hypoxia group, and DMSO control group were smaller at PSH 12 and 24 (with P values below 0.05), and the residual width of scratch of cells in HIF-1α stabilizer group was smaller at PSH 12 (P<0.05). Compared with those of cells in pure hypoxia group, the residual widths of scratch of cells in hypoxia group were wider at PSH 12 and 24 (with P values below 0.05), and the residual width of scratch of cells in HIF-1α inhibitor group was wider at PSH 12 (P<0.05), and those of cells in HIF-1α stabilizer group were smaller at PSH 12 and 24 (with P values below 0.05). There was no obvious difference in the width of scratch in cells among the 7 groups (F=19.02, P>0.05). The protein levels of HIF-1α in ESCs of wild-type mice at PHH 0, 12, 24, and 48 were respectively 1.02±0.05, 2.56±0.09, 1.60±0.17, and 1.17±0.03. Compared with that at PHH 0, the protein level of HIF-1α at PHH 12 was significantly enhanced (P<0.01). It began to decline at PHH 24 but was still higher than that at PHH 0 (P<0.05), and the protein level of HIF-1α at PHH 48 was close to the normoxia level (P>0.05). Compared with those of cells in pure hypoxia group, the mRNA expressions of P311 of cells in HIF-1α inhibitor group were significantly decreased at each time point (with P values below 0.05), and those in HIF-1α stabilizer group were significantly increased at PHH 12 and 24 (with P values below 0.05). Compared with those of cells in HIF-1α inhibitor group, the mRNA expressions of P311 of cells in DMSO control group and HIF-1α stabilizer group were significantly increased at PHH 0, 12, and 24 (with P values below 0.05). Compared with those of cells in pure hypoxia group, the expressions of P311 of cells in HIF-1α inhibitor group were significantly decreased at each time point (with P values below 0.05), while those in HIF-1α stabilizer group were significantly increased at PHH 12 and 24 (with P values below 0.05). Compared with those of cells in HIF-1α inhibitor group, the expressions of P311 of cells in DMSO control group and HIF-1α stabilizer group were significantly increased at PHH 12 and 24 (with P values below 0.05). (2) The results of HEK-293 cells. At PCH 0, there was no significant difference in the luciferase activity among cells of empty vector hypoxia group, P311 normoxia group, P311 hypoxia group, and P311 hypoxia+ HIF-1α inhibitor group (F=13.33, P>0.05). At PCH 12, the luciferase activity of cells in P311 hypoxia group was higher than that in empty vector hypoxia group (P<0.01). The luciferase activity of cells in hypoxia group was higher than that in P311 normoxia group (P<0.05), while that of cells in P311 hypoxia+ HIF-1α inhibitor group was lower than that in P311 hypoxia group (P<0.01). Conclusions: HIF-1α may increase the migration of murine ESCs through inducing the expression of P311 at the early stage of hypoxia.


Assuntos
Queimaduras/metabolismo , Células Epiteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia , Animais , Western Blotting , Hipóxia Celular , Movimento Celular , Proliferação de Células , Células Epiteliais/citologia , Vetores Genéticos , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso , Plasmídeos , Células-Tronco/citologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA