Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Adv Mater ; : e2405290, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39011814

ABSTRACT

In an emergency, nonvariceal upper gastrointestinal bleeding (NVUGIB), endoscopic hemostasis is considered the gold standard intervention. However, current endoscopic hemostasis is very challenging to manage bleeding in large-diameter or deep lesions highly prone to rebleeding risk. Herein, a novel hemostatic peptide hydrogel (HPH) is reported, consisting of a self-assembly peptide sequence CFLIVIGSIIVPGDGVPGDG (PFV) and gelatin methacryloyl (GelMA), which can be triggered by blue laser endoscopy (BLE) for nonvariceal upper gastrointestinal bleeding treatment without recurring bleeding concerns. Upon contact with GelMA solution, PFV immediately fibrillates into ß-sheet nanofiber and solvent-induced self-assembly to form HPH gel. HPH nanofiber networks induced ultrafast coagulation by enveloping blood cells and activating platelets and coagulation factors even to the blood with coagulopathy. Besides its remarkable hemostatic performance in artery and liver injury models, HPH achieves instant bleeding management in porcine NVUGIB models within 60 s by preventing the rebleeding risk. This work demonstrates an extraordinary hemostatic agent for NVUGIB intervention by BLE for the first time, broadening potential application scenarios, including patients with coagulopathy and promising clinical prospects.

2.
Elife ; 122023 May 25.
Article in English | MEDLINE | ID: mdl-37227051

ABSTRACT

The transition metal iron plays a crucial role in living cells. However, high levels of iron are potentially toxic through the production of reactive oxygen species (ROS), serving as a deterrent to the commensal fungus Candida albicans for colonization in the iron-rich gastrointestinal tract. We observe that the mutant lacking an iron-responsive transcription factor Hap43 is hyper-fit for colonization in murine gut. We demonstrate that high iron specifically triggers multiple post-translational modifications and proteasomal degradation of Hap43, a vital process guaranteeing the precision of intestinal ROS detoxification. Reduced levels of Hap43 de-repress the expression of antioxidant genes and therefore alleviate the deleterious ROS derived from iron metabolism. Our data reveal that Hap43 functions as a negative regulator for oxidative stress adaptation of C. albicans to gut colonization and thereby provide a new insight into understanding the interplay between iron homeostasis and fungal commensalism.


Subject(s)
Fungal Proteins , Iron , Animals , Mice , Iron/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Reactive Oxygen Species/metabolism , Candida albicans/genetics , Gastrointestinal Tract/microbiology , Homeostasis , Gene Expression Regulation, Fungal
3.
Oncogene ; 42(14): 1144-1156, 2023 03.
Article in English | MEDLINE | ID: mdl-36823376

ABSTRACT

Although accumulating evidence has highlighted the molecular mechanisms by which hTERT promotes tumour cell invasion and metastasis, the molecular mechanisms of the properties enabling hTERT to contribute to invasion and metastasis have not been clearly illustrated. Here, we report that hTERT promotes gastric cancer invasion and metastasis by recruiting p50 to synergistically inhibit PLEKHA7 expression. We observed that the expression of PLEKHA7 in gastric cancer was significantly negatively associated with the TNM stage and lymphatic metastasis and that decreased PLEKHA7 expression dramatically increased invasion and metastasis in gastric cancer cells. Further mechanistic research showed that hTERT directly regulates PLEKHA7 expression by binding p50 and recruiting the hTERT/p50 complex to the PLEKHA7 promoter. Increased hTERT dramatically decreased PLEKHA7 expression and promoted invasion and metastasis in gastric cancer cells. The hTERT-mediated invasion/metastasis properties at least partially depended on PLEKHA7. Our work uncovers a novel molecular mechanism underlying invasion/metastasis in gastric cancer orchestrated by hTERT and p50.


Subject(s)
Carrier Proteins , Stomach Neoplasms , Telomerase , Humans , Carrier Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Lymphatic Metastasis , Neoplasm Invasiveness , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Telomerase/genetics , Telomerase/metabolism
4.
FEBS Open Bio ; 10(11): 2316-2328, 2020 11.
Article in English | MEDLINE | ID: mdl-32935470

ABSTRACT

Acute lung injury (ALI) is accompanied by overactivation of multiple pro-inflammatory factors. Cytochrome P450 1A1 (CYP1A1) has been shown to aggravate lung injury in response to hyperoxia. However, the relationship between CYP1A1 and lipopolysaccharide (LPS)-induced ALI is unknown. In this study, CYP1A1 was shown to be upregulated in mouse lung in response to LPS. Using CYP1A1-deficient (CYP1A1-/-) mice, we found that CYP1A1 knockout enhanced LPS-induced ALI, as evidenced by increased TNF-α, IL-1ß, IL-6, and nitric oxide in lung; these effects were mediated by overactivation of NF-κB and iNOS. Furthermore, we found that aspartate aminotransferase, lactate dehydrogenase, creatine kinase, and creatinine levels were elevated in serum of LPS-induced CYP1A1-/- mice. Altogether, these data provide novel insights into the involvement of CYP1A1 in LPS-induced lung injury.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Cytochrome P-450 CYP1A1/genetics , NF-kappa B/metabolism , Acute Lung Injury/complications , Acute Lung Injury/pathology , Animals , Cytochrome P-450 CYP1A1/deficiency , Inflammation Mediators/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Aryl Hydrocarbon/metabolism , Sepsis/complications , Sepsis/pathology
5.
Cell Commun Signal ; 18(1): 74, 2020 May 18.
Article in English | MEDLINE | ID: mdl-32423412

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

6.
Cell Commun Signal ; 18(1): 70, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366266

ABSTRACT

The hydroxylase cytochrome P450 1A1 (CYP1A1) is regulated by the inflammation-limiting aryl hydrocarbon receptor (AhR), but CYP1A1 immune functions remain unclear. We observed CYP1A1 overexpression in peritoneal macrophages (PMs) isolated from mice following LPS or heat-killed Escherichia. coli (E. coli) challenge. CYP1A1 overexpression augmented TNF-α and IL-6 production in RAW264.7 cells (RAW) by enhancing JNK/AP-1 signalling. CYP1A1 overexpression also promoted 12S-hydroxy-5Z,8Z,10E,14Z-eicosatetraenoic acid (12(S)-HETE) production in activated RAW, while a 12(S)-HETE antibody attenuated and 12(S)-HETE alone induced inflammatory responses. Macrophages harbouring hydroxylase-deficient CYP1A1 demonstrated reduced 12(S)-HETE generation and LPS-induced TNF-α/IL-6 secretion. CYP1A1 overexpression also impaired phagocytosis of bacteria via decreasing the expression of scavenger receptor A (SR-A) in PMs. Mice injected with CYP1A1-overexpressing PMs were more susceptible to CLP- or E. coli-induced mortality and bacteria invading, while Rhapontigenin, a selective CYP1A1 inhibitor, improved survival and bacteria clearance of mice in sepsis. CYP1A1 and 12(S)-HETE were also elevated in monocytes and plasma of septic patients and positively correlated with SOFA scores. Macrophage CYP1A1 disruption could be a promising strategy for treating sepsis. Video abstract.


Subject(s)
Cytochrome P-450 CYP1A1/physiology , MAP Kinase Kinase 4/metabolism , Macrophages, Peritoneal , Phagocytosis , Sepsis/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Adult , Aged , Animals , Escherichia coli , Humans , Inflammation , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , RAW 264.7 Cells , Young Adult
8.
Cell Immunol ; 349: 104047, 2020 03.
Article in English | MEDLINE | ID: mdl-32019673

ABSTRACT

The polarization of macrophages is critical to inflammation and tissue repair, with unbalanced macrophage polarization associated with critical dysfunctions of the immune system. Cytochrome P450 1A1 (CYP1A1) is a hydroxylase mainly controlled by the inflammation-limiting aryl hydrocarbon receptor (AhR), which plays a critical role in mycoplasma infection, oxidative stress injury, and cancer. Arginase-1 (Arg-1) is a surrogate for polarized alternative macrophages and is important to the production of nitric oxide (NO) by the modulation of arginine. In the present study, we found CYP1A1 to be upregulated in IL-4-stimulated mouse peritoneal macrophages (PMs) and human peripheral blood monocytes. Using CYP1A1-overexpressing RAW264.7 cells (CYP1A1/RAW) we found that CYP1A1 augmented Arg-1 expression by strengthening the activation of the JAK1/STAT6 signaling pathway in macrophages treated with IL-4. 15(S)-HETE, a metabolite of CYP1A1 hydroxylase, was elevated in IL-4-induced CYP1A1/RAW cells. Further, in macrophages, the loss-of-CYP1A1-hydroxylase activity was associated with reduced IL-4-induced Arg-1 expression due to impaired 15(S)-HETE generation. Of importance, CYP1A1 overexpressing macrophages reduced the inflammation associated with LPS-induced peritonitis. Taken together, these findings identified a novel signaling axis, CYP1A1-15(S)-HETE-JAK1-STAT6, that may be a promising target for the proper maintenance of macrophage polarization and may also be a means by which to treat immune-related disease due to macrophage dysfunction.


Subject(s)
Arginase/biosynthesis , Cytochrome P-450 CYP1A1/physiology , Janus Kinase 1/antagonists & inhibitors , Macrophages, Peritoneal/drug effects , Peritonitis/prevention & control , STAT6 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Adoptive Transfer , Animals , Arachidonate 15-Lipoxygenase/physiology , Arginase/genetics , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Endotoxins/toxicity , Humans , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/genetics , Hydroxyeicosatetraenoic Acids/pharmacology , Interleukin-4/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/transplantation , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Peritonitis/chemically induced , RAW 264.7 Cells , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , THP-1 Cells , Up-Regulation/drug effects
9.
Cytokine ; 128: 155001, 2020 04.
Article in English | MEDLINE | ID: mdl-32035329

ABSTRACT

Neutrophilic granule protein (NGP) belongs to the cystatin superfamily. Even though this superfamily is critically involved in cancer biology and adaptive immunity, the relationship of macrophage NGP to inflammation and phagocytosis remains poorly understood. In this study, we observed a significant increase of NGP in peritoneal macrophages (PMs) isolated from mice challenged with E. coli or lipopolysaccharide (LPS), as judged by NGP mRNA microarray. We also found changes in NGP to be mainly Toll-like receptor 4 (TLR4)-dependent. By western blot and electrophoretic mobility shift assay, we demonstrated NGP overexpression to reduce TNF-α and IL-1ß production by LPS-induced RAW264.7 cells (RAW) via suppression of the NF-κB (p65 and p50) signalling pathway, rather than the JNK1/AP-1 (fos and jun) signalling pathway. NGP overexpression by LPS-induced RAW also induced IL-10, an anti-inflammatory cytokine, which was partially involved in the anti-inflammatory effect produced by NGP overexpression. Moreover, upregulated NGP enhanced the phagocytosis of E. coli by RAW. Taken together, these results demonstrated NGP to be an important host defense component that regulates inflammatory responses and phagocytosis by activated macrophages. As such, NGP may be useful for the treatment of inflammatory based disease.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Phagocytosis/physiology , Animals , Cell Line , Cystatins/metabolism , Cytokines/metabolism , Escherichia coli/metabolism , Inflammation/pathology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Inflammation ; 43(1): 231-240, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31802382

ABSTRACT

Ellipticine, a natural product from Ochrosia elliptica, has been broadly investigated for its anticancer effects. Although inflammation has been clearly identified as a key factor in the onset and progression of cancer, the relationship between ellipticine and inflammation remains unknown. Hence, the aims of the present study were to assess the effects of ellipticine on the inflammatory responses to lipopolysaccharide (LPS)-induced macrophages and to potentially identify the underlying mechanisms involved. Viability testing showed that ellipticine was not significantly toxic to Raw264.7 cells and actually conveyed protective effects to LPS-stimulated Raw264.7 cells and human peripheral blood monocytes by decreasing the secretion of inflammatory factors (TNF-α and IL-6). The results of western blot analysis and electrophoretic mobility shift assays showed that ellipticine markedly suppressed LPS-induced activation of the JNK/AP-1 (c-Fos and c-Jun) signaling pathway, but not ERK/p38/NF-κB pathway (p65 and p50) activation. Furthermore, ellipticine reduced the inflammatory response and mortality in a mouse model of LPS-induced endotoxic shock. Collectively, these data indicate that ellipticine may be a potential therapeutic agent for the treatment of inflammation-associated diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ellipticines/pharmacology , Inflammation/prevention & control , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Shock, Septic/prevention & control , Transcription Factor AP-1/metabolism , Adult , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/immunology , Interleukin-6/metabolism , Lipopolysaccharides , Macrophages/enzymology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Shock, Septic/chemically induced , Shock, Septic/enzymology , Shock, Septic/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
11.
Crit Care Med ; 48(1): e40-e47, 2020 01.
Article in English | MEDLINE | ID: mdl-31634234

ABSTRACT

OBJECTIVES: The knowledge that agmatine is found in the human body has existed for several years; however, its role in sepsis has not yet been studied. In the present study, we investigate the role of agmatine in the progression and treatment of sepsis. DESIGN: Clinical/laboratory investigations. SETTING: Medical centers/University-based research laboratory. SUBJECTS: Elective ICU patients with severe sepsis and healthy volunteers; C57BL/6 mice weighing 18-22 g. INTERVENTIONS: Serum agmatine level and its associations with inflammatory markers were assessed in patients with sepsis. Agmatine was administered intraperitoneally to mice before a lipopolysaccharide challenge. Human peripheral blood mononuclear cells and murine macrophages were pretreated with agmatine followed by lipopolysaccharide stimulation. MEASUREMENTS AND MAIN RESULTS: Serum agmatine levels were significantly decreased in patients with sepsis and lipopolysaccharide-induced mice, and correlated with Acute Physiology and Chronic Health Evaluation II score, procalcitonin, tumor necrosis factor-α, and interleukin-6 levels. In a therapeutic experiment, exogenous agmatine attenuated the cytokine production of peripheral blood mononuclear cells from patients with sepsis and healthy controls. Agmatine also exerted a significant beneficial effect in the inflammatory response and organ damage and reduced the death rate in lipopolysaccharide-induced mice. Imidazoline I2 receptor agonist 2-benzofuran-2-yl blocked the pharmacological action of agmatine; whereas, other imidazoline receptor ligands did not. Furthermore, agmatine significantly impaired the inflammatory response by inactivating nuclear factor-κB, but not protein 38 mitogen-activated protein kinase, c-Jun N-terminal kinase, extracellular signal-regulated kinase, and inducible nitric oxide synthase signaling in macrophages. Activation of imidazoline I2 receptor or knockdown of ribosomal S6 kinase 2 counteracted the effects of agmatine on phosphorylation and degradation of inhibitor of nuclear factor-κBα. CONCLUSIONS: Endogenous agmatine metabolism correlated with the progression of sepsis. Supplemental exogenous agmatine could ameliorate the lipopolysaccharide-induced systemic inflammatory responses and multiple organ injuries through the imidazoline I2 receptor-ribosomal S6 kinase 2-nuclear factor-κB pathway. Agmatine could be used as both a clinical biomarker and a promising pharmaconutrient in patients with severe sepsis.


Subject(s)
Agmatine/therapeutic use , Imidazoline Receptors/physiology , NF-kappa B/physiology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Sepsis/drug therapy , Signal Transduction/physiology , Agmatine/pharmacology , Animals , Cells, Cultured , Disease Progression , Humans , Imidazoline Receptors/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred C57BL , NF-kappa B/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/drug effects , Signal Transduction/drug effects
12.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 31(9): 1163-1166, 2019 Sep.
Article in Chinese | MEDLINE | ID: mdl-31657346

ABSTRACT

OBJECTIVE: Infection is one of the main causes of death in clinical patients, and multi-drug resistance leads to ineffective treatment with conventional antibiotics. Therefore, it is imperative to develop new anti-infective drugs. Antimicrobial peptides cathelicidins are cationic host defense peptides found in many organisms. It has been demonstrated by in vivo and in vitro studies that antimicrobial peptides cathelicidins not only show broad-spectrum antibacterial activity and high sensitivity to drug-resistant bacteria, but also have a good guiding effect on the immune response. This paper summarizes the reports of antimicrobial peptides cathelicidins in recent years, highlighting their research achievements in antibiosis, anti-inflammatory, chemotaxis regulation and phagocytosis, providing new ideas for the treatment of infection-related diseases.


Subject(s)
Anti-Infective Agents , Antimicrobial Cationic Peptides , Cathelicidins , Anti-Bacterial Agents , Bacteria , Humans
13.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 31(6): 777-780, 2019 Jun.
Article in Chinese | MEDLINE | ID: mdl-31315741

ABSTRACT

OBJECTIVE: Infectious and inflammatory diseases are important diseases threatening human health. Without timely control, a series of complications will occur in patients, such as sepsis, inflammatory factor storm, and even lead to death. It has been found that cytochrome P4501A1 (CYP1A1) plays a key role in the development of infectious and inflammatory diseases through aromatic hydrocarbon receptor (AhR) dependent and non-dependent pathways in different cells and organs induced by different substances. The non AhR dependent regulatory mechanism of CYP1A1 and the different roles of CYP1A1 in infection and inflammation is reviewed in order to provide reference for further research on the relationship between CYP1A1 and infection and inflammation.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Infections/etiology , Inflammation/etiology , Receptors, Aryl Hydrocarbon/metabolism , Humans
14.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 31(12): 1555-1558, 2019 Dec.
Article in Chinese | MEDLINE | ID: mdl-32029051

ABSTRACT

OBJECTIVE: 12-HETE is a metabolite of arachidonic acid (AA). AA is normally present in membrane phospholipids. The exposure to different stimuli can trigger the release of AA through the activity of phospholipase A2 (PLA2) by cells. An important metabolic pathway which utilizes AA as its substrate is 12-Lipoxygenase (12-LOX), resulting in the formation of 12-HETE. 12-HETE plays an important role in many diseases such as cancer, diabetes, hypertension, and participates in the pathogenesis of inflammation and oxidative stress and other pathological processes.Current research shows that it participates in metamorphism and exudation in the process of inflammation. This review is aimed at summarizing its role in inflammation and oxidative stress, with improved understanding of 12-HETE.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Oxidative Stress , Arachidonic Acid , Humans , Inflammation
15.
Front Immunol ; 9: 2033, 2018.
Article in English | MEDLINE | ID: mdl-30283437

ABSTRACT

The aryl hydrocarbon receptor (AhR) is an important immune regulator with a role in inflammatory response. However, the role of AhR in IL-10 production by inflammatory macrophages is currently unknown. In this study, we investigated LPS-induced IL-10 expression in macrophages from AhR-KO mice and AhR-overexpressing RAW264.7 cells. AhR was highly expressed after LPS stimulation through NF-κB pathway. Loss of AhR resulted in reduced IL-10 expression in LPS-induced macrophages. Moreover, the IL-10 expression was elevated in LPS-induced AhR-overexpressing RAW264.7 cells. Maximal IL-10 expression was dependent on an AhR non-genomic pathway closely related to Src and STAT3. Furthermore, AhR-associated Src activity was responsible for tyrosine phosphorylation of STAT3 and IL-10 expression by inflammatory macrophages. Adoptive transfer of AhR-expressing macrophages protected mice against LPS-induced peritonitis associated with high IL-10 production. In conclusion, we identified the AhR-Src-STAT3-IL-10 signaling pathway as a critical pathway in the immune regulation of inflammatory macrophages, It suggests that AhR may be a potential therapeutic target in immune response.


Subject(s)
Inflammation/immunology , Interleukin-10/immunology , Macrophages/immunology , Receptors, Aryl Hydrocarbon/immunology , STAT3 Transcription Factor/immunology , src-Family Kinases/immunology , Adoptive Transfer , Animals , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/immunology , Peritonitis/metabolism , Peritonitis/therapy , RAW 264.7 Cells , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
16.
Mil Med Res ; 3: 33, 2016.
Article in English | MEDLINE | ID: mdl-27833759

ABSTRACT

Trauma still represents one of the major causes of death worldwide. Despite the reduction of post-traumatic sepsis over the past two decades, the mortality of septic trauma inpatients is still high (19.5-23 %). Early prevention of sepsis development can aid in the subsequent treatment of patients and help improve their outcomes. To date, the prevention of trauma-related infection/sepsis has mainly included infection prevention (e.g., surgical management, prophylactic antibiotics, tetanus vaccination, immunomodulatory interventions) and organ dysfunction prevention (e.g., pharmaceuticals, temporary intravascular shunts, lung-protective strategies, enteral immunonutrition, acupuncture). Overall, more efficient ways should be developed to prevent trauma-related infection/sepsis.


Subject(s)
Multiple Trauma/complications , Sepsis , Wound Infection , Humans , Sepsis/etiology , Sepsis/prevention & control , Wound Infection/etiology , Wound Infection/prevention & control
17.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 28(8): 761-4, 2016 Aug.
Article in Chinese | MEDLINE | ID: mdl-27434574

ABSTRACT

OBJECTIVE: High mobility group protein B1 (HMGB1) is the most representative substance in the alarmins family, it is actively or passively release to extracellular by the activation of monocyte/macrophage and the dead cells, and then it stimulates the production of a variety of inflammatory mediators, and increases the organism's inflammatory response through relevant receptors signaling pathways. In recent years, its concentration can reflect the severity of inflammation and injury and was related to the prognosis, HMGB1 has won more and more attention in the development of sepsis. By reviewing the study of HMGB1 in sepsis pathogenesis, signal pathway and reversal measures, it was found that HMGB1 was considered as an important inflammatory mediators and warning signal involved in the pathogenesis of sepsis, and was become a new target in the treatment of sepsis. Further research on the role of HMGB1 in the pathogenesis of sepsis is needed in the future, and the development of new drugs combined with HMGB1 will be used in the study of HMGB1 in animal experiments.


Subject(s)
Alarmins/metabolism , HMGB1 Protein/metabolism , Sepsis/metabolism , Signal Transduction , Animals , Humans , Inflammation/metabolism , Macrophages/metabolism , Monocytes/metabolism
18.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 28(3): 225-9, 2016 Mar.
Article in Chinese | MEDLINE | ID: mdl-29916686

ABSTRACT

Objective: To investigate the protective effect of agmatine (AGM) against peritoneal inflammatory response and neutrophil (PMN) infiltration induced by zymosan (ZYM) in mice. Methods: Thirty-six adult male C57BL/6 mice were randomly divided into sham group, model group, and AGM treatment group. Peritonitis model was reproduced by intra-peritoneal injection of 1 mg/mL ZYM (0.5 mL), while equivalent phosphate buffer saline (PBS) was given to sham group. 200 mg/kg AGM was injected into peritoneal cavity after ZYM challenge in AGM treatment group. Six mice in each group were sacrificed at 2 hours and 6 hours, respectively, after reproduction of the model. Blood sample and peritoneal lavage fluid (PLF) were collected. The levels of keratinocyte-derived chemokine (KC), macrophage inflammatory protein 2 (MIP-2), tumor necrosis factor-α (TNF-α), interleukins-6 (IL-6) in serum and PLF were determined by enzyme linked immunosorbent assay (ELISA). The number of leukocytes and PMN in PLF were determined by hemocytometer and flow cytometry, respectively. Results: Compared with sham group, all serum and PLF levels of KC, MIP-2, TNF-α and IL-6 were greatly elevated at 2 hours after ZYM injection in model group, while AGM treatment could dramatically reduce the levels of the above-mentioned cytokines in serum and PLF as compared with those of the model group [serum KC (ng/L): 990.7±137.9 vs. 2 053.2±262.7, MIP-2 (ng/L): 642.2±124.4 vs. 1 369.7±146.5, TNF-α (ng/L): 608.6±38.1 vs. 1 044.7±101.0, IL-6 (ng/L): 1 058.2±129.1 vs. 1 443.3±190.1; PLF KC (ng/L): 7 462.3±839.6 vs. 12 723.5±1 515.7, MIP-2 (ng/L): 1 570.8±193.4 vs. 3 471.4±384.7, TNF-α (ng/L): 1 115.8±156.7 vs. 1 499.2±231.2, IL-6 (ng/L): 2 646.5±223.2 vs. 3 126.7±291.4; all P < 0.05]. The expressions of KC, MIP-2 and TNF-α at 6 hours were significantly lower than those at 2 hours in model group and AGM treatment group, but IL-6 levels were further increased. The levels of KC and MIP-2 in serum and PLF at 6 hours were decreased to the levels of sham group. At 6 hours after the reproduction of the model, the number of total inflammatory cells and PMN of PLF in the model group was significantly higher than those of the sham group. In contrast, AGM notably lowered the number of inflammatory cells and PMN in peritoneal fluid after ZYM attack [total inflammatory cells (×109/L): 14.7±1.1 vs. 2.0±0.4, 10.1±1.2 vs. 14.7±1.1; PMN (×109/L): 11.37±1.22 vs. 0.18±0.05, 7.69±0.57 vs. 11.37±1.22, all P < 0.05]. Conclusion: AGM can effectively alleviate acute peritoneal inflammatory injury induced by ZYM, mainly through reducing the secretion of inflammatory mediators and chemokines, and inhibiting the infiltration of leukocytes and neutrophils.


Subject(s)
Agmatine/pharmacology , Cytokines/metabolism , Inflammation/drug therapy , Neutrophil Infiltration/drug effects , Peritoneal Diseases/immunology , Zymosan/pharmacology , Animals , Chemokines/metabolism , Interleukin-6 , Male , Mice , Mice, Inbred C57BL , Neutrophils , Peritoneal Diseases/drug therapy , Random Allocation , Tumor Necrosis Factor-alpha
19.
Cell Transplant ; 24(5): 891-907, 2015.
Article in English | MEDLINE | ID: mdl-24835482

ABSTRACT

Genetically engineered stem cells that overexpress genes encoding therapeutic products can be exploited to correct metabolic disorders by repairing and regenerating diseased organs or restoring their function. Hair follicles are readily accessible and serve as a rich source of autologous stem cells for cell-based gene therapy. Here we isolated mesenchymal stem cells from human hair follicles (HF-MSCs) and engineered them to overexpress the human insulin gene and release human insulin in a time- and dose-dependent manner in response to rapamycin. The engineered HF-MSCs retained their characteristic cell surface markers and retained their potential to differentiate into adipocytes and osteoblasts. When mice with streptozotocin-induced type 1 diabetes were engrafted with these engineered HF-MSCs, these cells expressed and released a dose of human insulin, dramatically reversed hyperglycemia, and significantly reduced death rate. Moreover, the engineered HF-MSCs did not form detectable tumors throughout the 120-day animal tests in our experiment. Our results show that HF-MSCs can be used to safely and efficiently express therapeutic transgenes and therefore show promise for cell-based gene therapy of human disease.


Subject(s)
Cell Engineering , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Hair Follicle/metabolism , Hyperglycemia/therapy , Insulin/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Adult , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , Heterografts , Humans , Hyperglycemia/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL