Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Clin Sci (Lond) ; 135(22): 2541-2558, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34730176

ABSTRACT

OBJECTIVE: Regulated in development and DNA damage responses-1 (REDD1) is a conserved and ubiquitous protein, which is induced in response to multiple stimuli. However, the regulation, function and clinical relevance of REDD1 in Helicobacter pylori-associated gastritis are presently unknown. APPROACH: Immunohistochemistry, real-time PCR and Western blot analyses were performed to examine the levels of REDD1 in gastric samples from H. pylori-infected patients and mice. Gastric tissues from Redd1-/- and wildtype (WT, control) mice were examined for inflammation. Gastric epithelial cells (GECs), monocytes and T cells were isolated, stimulated and/or cultured for REDD1 regulation and functional assays. RESULTS: REDD1 was increased in gastric mucosa of H. pylori-infected patients and mice. H. pylori induced GECs to express REDD1 via the phosphorylated cytotoxin associated gene A (cagA) that activated MAPKp38 pathway to mediate NF-κB directly binding to REDD1 promoter. Human gastric REDD1 increased with the severity of gastritis, and mouse REDD1 from non-marrow chimera-derived cells promoted gastric inflammation that was characterized by the influx of MHCII+ monocytes. Importantly, gastric inflammation, MHCII+ monocyte infiltration, IL-23 and IL-17A were attenuated in Redd1-/- mice. Mechanistically, REDD1 in GECs regulated CXCL1 production, which attracted MHCII+ monocytes migration by CXCL1-CXCR2 axis. Then H. pylori induced MHCII+ monocytes to secrete IL-23, which favored IL-17A-producing CD4+ cell (Th17 cell) polarization, thereby contributing to the development of H. pylori-associated gastritis. CONCLUSIONS: The present study identifies a novel regulatory network involving REDD1, which collectively exert a pro-inflammatory effect within gastric microenvironment. Efforts to inhibit this REDD1-dependent pathway may prove valuable strategies in treating of H. pylori-associated gastritis.


Subject(s)
Cytokines/metabolism , Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Th17 Cells/microbiology , Transcription Factors/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Case-Control Studies , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastritis/immunology , Gastritis/metabolism , Helicobacter Infections/complications , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Host-Pathogen Interactions , Humans , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phenotype , Phosphorylation , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
2.
FASEB J ; 34(1): 1169-1181, 2020 01.
Article in English | MEDLINE | ID: mdl-31914631

ABSTRACT

BHLHE40, a member of the basic helix-loop-helix transcription factor family, has been reported to play an important role in inflammatory diseases. However, the regulation and function of BHLHE40 in Helicobacter pylori (H pylori)-associated gastritis is unknown. We observed that gastric BHLHE40 was significantly elevated in patients and mice with H pylori infection. Then, we demonstrate that H pylori-infected GECs express BHLHE40 via cagA-ERK pathway. BHLHE40 translocates to cell nucleus, and then binds to cagA protein-activated p-STAT3 (Tyr705). The complex increases chemotactic factor CXCL12 expression (production). Release of CXCL12 from GECs fosters CD4+ T cell infiltration in the gastric mucosa. Our results identify the cagA-BHLHE40-CXCL12 axis that contributes to inflammatory response in gastric mucosa during H pylori infection.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Chemokine CXCL12/metabolism , Epithelial Cells/metabolism , Gastritis/microbiology , Helicobacter Infections/metabolism , Homeodomain Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Nucleus/metabolism , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastritis/metabolism , Gene Expression Regulation , Helicobacter pylori , Humans , Inflammation , Mice , Mice, Inbred C57BL , Stomach/microbiology , Up-Regulation
3.
FASEB J ; 33(4): 5018-5033, 2019 04.
Article in English | MEDLINE | ID: mdl-30596522

ABSTRACT

Cathepsin C (CtsC) functions as a central coordinator for activation of many serine proteases in immune cells. However, CtsC expression in gastric epithelial cells and its role in Helicobacter pylori infection remain unclear. Real-time PCR, Western blot, and immunohistochemistry analyses identified that CtsC was decreased in gastric mucosa of H. pylori-infected patients and mice. Isolated gastric epithelial cells and cell lines were stimulated with H. pylori and/or TGF-Ɵ1 showed that down-regulation of CtsC in gastric epithelial cells largely depended on H. pylori cagA via Src/ERK and Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathways, and the effect could be synergistically augmented by TGF-Ɵ1 in an autocrine manner. In human gastric mucosa, CtsC expression was negatively correlated with bacteria colonization; accordingly, provision of exogenous active CtsC overwhelmed H. pylori persistence in gastric mucosa of mice. In the presence of active CtsC, isolated human neutrophils activated via NF-κB pathway with augmented bactericidal capacity in vitro. We also found that neutrophils activated and cleared bacteria in active CtsC-injected mice and that there was no bactericidal capacity in mice that were simultaneously neutrophil-depleted by Ly6G antibody. Our findings identified a mechanism that H. pylori abrogate CtsC to impair neutrophil activation and to ensure persistence in gastric mucosa. Efforts to enable and boost this neutrophil activation pathway by active CtsC may therefore become valuable strategies in treating H. pylori infection.-Liu, Y. G., Teng, Y. S., Cheng, P., Kong, H., Lv, Y. P., Mao, F. Y., Wu, X. L., Hao, C. J., Chen, W., Yang, S. M., Zhang, J. Y., Peng, L. S., Wang, T. T., Han, B., Ma, Q., Zou, Q. M., Zhuang, Y. Abrogation of cathepsin C by Helicobacter pylori impairs neutrophil activation to promote gastric infection.


Subject(s)
Cathepsin C/metabolism , Gastric Mucosa/microbiology , Helicobacter pylori/pathogenicity , Neutrophil Activation/physiology , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Helicobacter Infections/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Phagocytosis/physiology , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
4.
Eur Heart J ; 35(20): 1327-34, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24497338

ABSTRACT

AIM: Although catheter ablation (CA) has replaced antiarrhythmic drugs (AAD) as first-line treatment in selected patients with atrial fibrillation (AF), optimal treatment of recurrent atrial tachycardia (AT) after AF ablation remains unclear. This parallel randomized controlled study compared CA vs. AAD for recurrent AT after persistent AF ablation. METHODS AND RESULTS: Two-hundred and one patients (aged 59.1 Ā± 10.9 years, 68.7% male) with recurrent AT after persistent AF ablation were enrolled and randomized to either CA (n = 101) or AAD (n = 100) treatment. Primary endpoint was freedom from recurrent atrial tachyarrhythmia (ATa, including AT and AF) at 24-month follow-up. Composite secondary endpoints comprised procedural complications, long-term morbidity and improvement in quality of life (QoL). On an intention-to-treat basis, the CA group had a higher rate of freedom from recurrent ATa (56.4 vs. 34.0%; P = 0.001). Adjusted Cox regression analysis showed a significant treatment effect with a hazard ratio of 0.538 (95% CI: 0.355-0.816) in favour of CA. There was a higher proportion of periprocedural complications in the CA group (7.9 vs. 0; P = 0.012), and of long-term adverse events in the AAD group (10.9 vs. 24.0%; P = 0.014). Quality of life was significantly higher for CA. CONCLUSIONS: This study demonstrates superiority of CA over AAD for recurrent AT after persistent AF ablation with regard to SR maintenance, long-term safety and QoL improvement. However, CA use might be limited by a higher risk for periprocedural complications.


Subject(s)
Atrial Fibrillation/surgery , Catheter Ablation/methods , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Atrial Fibrillation/mortality , Catheter Ablation/mortality , Chronic Disease , Electrophysiologic Techniques, Cardiac/methods , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prospective Studies , Quality of Life , Recurrence , Secondary Prevention/methods , Treatment Outcome
5.
Pacing Clin Electrophysiol ; 36(10): 1202-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23678857

ABSTRACT

BACKGROUND: This randomized prospective study compared three ablation strategies in patients with long-standing persistent atrial fibrillation (LPeAF). It also explored the best procedural endpoint from among the following: circumferential pulmonary vein isolation (PVI) + left atrial (LA) linear lesions (roofline, mitral isthmus) + complex fractionated atrial electrogram (CFAE) ablation, PVI + LA linear lesions + cavotricuspid isthmus (CTI) ablation + CFAE ablation, and PVI + CFAE ablation. METHODS AND RESULTS: A total of 210 patients with LPeAF referred for catheter ablation were enrolled and randomized into three ablation groups. The patients in group A (n = 70) underwent PVI followed by LA linear and CFAE ablation; in 93% of patients the primary endpoint was achieved (five patients with incomplete linear lesions). Of the 70 patients in group B who were subjected to PVI followed by LA linear, CFAE, and CTI ablations, in 94% of patients the primary endpoint was achieved (four patients with incomplete linear lesions). All patients in group C (n = 70) successfully underwent PVI and CFAE ablation. Direct current cardioversion was performed upon PVI, CFAE elimination, and completion of linear lesions. Patients were followed-up for atrial tachyarrhythmia recurrence for at least 24 months. After a single ablation procedure, group C (36%) exhibited the lowest success compared with group A (54%) and group B (51%) (P = 0.06). At the mean follow-up of 32 Ā± 9 months after the final ablation procedure, 53 patients (76%) in group A, 53 (76%) in group B, and 41 (59%) in group C were in sinus rhythm without antiarrhythmic drugs (P = 0.03). CONCLUSIONS: In LPeAF, linear lesions in the LA help improve outcome of ablation, additional CTI ablation does not.


Subject(s)
Atrial Fibrillation/epidemiology , Atrial Fibrillation/surgery , Catheter Ablation/methods , Catheter Ablation/statistics & numerical data , Heart Atria/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Atrial Fibrillation/diagnosis , China/epidemiology , Chronic Disease , Female , Humans , Male , Middle Aged , Prevalence , Risk Factors , Treatment Outcome , Young Adult
6.
Pacing Clin Electrophysiol ; 36(10): 1236-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23822135

ABSTRACT

OBJECTIVES: It is uncertain whether gender affects the outcomes of catheter ablation (CA) for atrial fibrillation (AF). The objective of the study is to evaluate the efficacy and safety of CA for long-standing persistent AF in women. METHODS: Between January 2010 and May 2011, 220 consecutive patients (73 females, 33.2%), with long-standing persistent AF who underwent CA were prospectively recruited. Gender-related differences in clinical presentation, periprocedural complications, and outcomes were compared. RESULTS: Women were less likely to have lone AF than men (27.4% vs 47.6%; P = 0.004). The incidence of rheumatic heart disease was higher in women (19.2% in women vs 1.4% in men; P < 0.001). Women had a lower initial ablation success rate than men (35.6% vs 57.1%; P = 0.003). Hematomas occurred more often in women (6.8% in women vs 0.7% in men; P = 0.027). A Cox regression analysis demonstrated total duration of AF (per month, hazard ratio [HR] 1.003, confidence interval [CI] 1.001-1.006; P = 0.006) and gender (HR 1.663, CI 1.114-2.485; P = 0.013) as the independent predictors for recurrence after the first CA. CONCLUSIONS: Women and long AF duration were closely related to the recurrence of AF after the first ablation in patients with long-standing persistent AF. Women also had a higher risk of vascular complications.


Subject(s)
Atrial Fibrillation/mortality , Atrial Fibrillation/surgery , Catheter Ablation/mortality , Postoperative Complications/mortality , Women's Health/statistics & numerical data , China/epidemiology , Chronic Disease , Comorbidity , Female , Humans , Male , Middle Aged , Prevalence , Risk Factors , Sex Distribution , Survival Rate , Treatment Outcome
7.
Zhonghua Yi Xue Za Zhi ; 93(31): 2505-7, 2013 Aug 20.
Article in Zh | MEDLINE | ID: mdl-24300277

ABSTRACT

OBJECTIVE: To evaluate the diagnostic significance of Fascin protein in non-small cell lung cancer (NSCLC) patients. METHODS: Among 110 cases of NSCLC patients and 50 cases of healthy controls, double antibody sandwich enzyme-linked immunosorbent assay (ELISA) was employed to measure the serum levels of Fascin protein, carcinoembryonic antigen (CEA), neuron-specific enolase (NSE), scales like squamous cell carcinoma (SCC) antigen, cytokeratin protein 21-1 (CYFRA21-1) and gastrin-releasing peptide (GRP) precursor. Fascin-positive rate (24.4%) in NSCLC patients was significantly higher than that in healthy controls (4%) (P < 0.05).In different clinical stages of non-small cell lung cancer patients, Fascin-positive rate was statistically significant (P < 0.05). The positive rates of Fascin for CEA, CYFRA21-1, NSE, SCC and Pro-GRP were 25.64%, 5.1%, 5.1%, 5.0% and 1.7% respectively. CONCLUSION: As a new tumor marker, Fascin may be used for clinical screening and prognosis in lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Carrier Proteins/blood , Lung Neoplasms/diagnosis , Microfilament Proteins/blood , Adult , Aged , Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/blood , Case-Control Studies , Female , Humans , Lung Neoplasms/blood , Male , Middle Aged
8.
Biochem Biophys Res Commun ; 411(3): 586-92, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21763284

ABSTRACT

Recent studies have revealed that microRNA-29c (miR-29c) is involved in a variety of biological processes including carcinogenesis. Here, we report that miR-29c was significantly downregulated in hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) cell lines as well as in clinical tissues compared with their corresponding controls. Tumor necrosis factor alpha-induced protein 3 (TNFAIP3), a key regulator in inflammation and immunity, was found to be inversely correlated with miR-29c levels and was identified as a target of miR-29c. Overexpression of miR-29c in HepG2.2.15 cells effectively suppressed TNFAIP3 expression and HBV DNA replication as well as inhibited cell proliferation and induced apoptosis. We conclude that miR-29c may play an important role as a tumor suppressive microRNA in the development and progression of HBV-related HCC by targeting TNFAIP3. Thus miR-29c and TNFAIP3 represent key diagnostic markers and potential therapeutic targets for the prevention and treatment of HBV infection.


Subject(s)
Carcinoma, Hepatocellular/virology , Gene Expression Regulation, Neoplastic , Hepatitis B virus , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/virology , MicroRNAs/biosynthesis , Nuclear Proteins/genetics , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins , Down-Regulation , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatitis B virus/physiology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Transfection , Tumor Necrosis Factor alpha-Induced Protein 3
9.
J Trauma ; 70(6): 1519-23, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21336201

ABSTRACT

BACKGROUND: Our objective was to observe the role of vascular endothelial growth factor (VEGF) 121 gene transfer in promoting vascular reconstruction and bone repair in femur head necrosis of rabbits. METHODS: The femoral head necrosis model was induced by injection with ethanol. The necrotic femoral head was transfected with a human adenoviral vector expressing VEGF (Ad-hVEGF121). Bone formation in the subchondral necrotic region was analyzed using histology, by measuring the bone mineral density value, and by observing bone trabecular morphology using image analysis. RESULTS: Revascularization level, bone formation rate, bone quality and quantity, and mineralization level in the subchondral necrotic region of the gene transfection group were significantly higher than the control groups. The control groups had more subchondral bone resorption compared with the gene transfection group. CONCLUSION: VEGF might promote bone formation and revascularization in the subchondral necrotic region of the femoral head, indirectly protecting the necrotic bone trabecula from absorption and avoiding a reduction in the mechanical function of the subchondral region.


Subject(s)
Femur Head Necrosis/drug therapy , Osteogenesis/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology , Absorptiometry, Photon , Adenoviridae , Analysis of Variance , Animals , Bone Density , Bone Resorption , Disease Models, Animal , Femur Head Necrosis/diagnostic imaging , Genetic Therapy/methods , Neovascularization, Physiologic/drug effects , Rabbits , Transfection
10.
Cell Mol Gastroenterol Hepatol ; 12(2): 395-425, 2021.
Article in English | MEDLINE | ID: mdl-33676046

ABSTRACT

BACKGROUND & AIMS: Rev-erbα represents a powerful transcriptional repressor involved in immunity. However, the regulation, function, and clinical relevance of Rev-erbα in Helicobacter pylori infection are presently unknown. METHODS: Rev-erbα was examined in gastric samples from HĀ pylori-infected patients and mice. Gastric epithelial cells (GECs) were isolated and infected with HĀ pylori for Rev-erbα regulation assays. Gastric tissues from Rev-erbα-/- and wild-type (littermate control) mice or these mice adoptively transferred with CD4+ T cells from IFN-ƎĀ³-/- and wild-type mice, bone marrow chimera mice and mice with inĀ vivo pharmacological activation or inhibition of Rev-erbα were examined for bacteria colonization. GECs, CD45+CD11c-Ly6G-CD11b+CD68- myeloid cells and CD4+ T cells were isolated, stimulated and/or cultured for Rev-erbα function assays. RESULTS: Rev-erbα was increased in gastric mucosa of HĀ pylori-infected patients and mice. HĀ pylori induced GECs to express Rev-erbα via the phosphorylated cagA that activated ERK signaling pathway to mediate NF-κB directly binding to Rev-erbα promoter, which resulted in increased bacteria colonization within gastric mucosa. Mechanistically, Rev-erbα in GECs not only directly suppressed Reg3b and Ɵ-defensin-1 expression, which resulted in impaired bactericidal effects against HĀ pylori of these antibacterial proteins inĀ vitro and inĀ vivo; but also directly inhibited chemokine CCL21 expression, which led to decreased gastric influx of CD45+CD11c-Ly6G-CD11b+CD68- myeloid cells by CCL21-CCR7-dependent migration and, as a direct consequence, reduced bacterial clearing capacity of HĀ pylori-specific Th1 cell response. CONCLUSIONS: Overall, this study identifies a model involving Rev-erbα, which collectively ensures gastric bacterial persistence by suppressing host gene expression required for local innate and adaptive defense against HĀ pylori.


Subject(s)
Adaptive Immunity , Helicobacter Infections/immunology , Helicobacter pylori/physiology , Host-Pathogen Interactions/immunology , Immunity, Innate , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Stomach/microbiology , Adult , Aged , Antigens, Bacterial/metabolism , Antigens, CD/metabolism , Bacterial Proteins/metabolism , Cell Movement , Colony Count, Microbial , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/blood , Helicobacter Infections/microbiology , Humans , MAP Kinase Signaling System , Male , Middle Aged , Models, Biological , Myeloid Cells/metabolism , NF-kappa B/metabolism , Pancreatitis-Associated Proteins/metabolism , Stomach/pathology , Th1 Cells/immunology , Young Adult , beta-Defensins/metabolism
11.
Mol Cancer Res ; 19(6): 968-978, 2021 06.
Article in English | MEDLINE | ID: mdl-33771880

ABSTRACT

Actin cytoskeleton dynamic rearrangement is required for tumor cell metastasis and is a key characteristic of Helicobacter pylori (H. pylori)-infected host cells. Actin cytoskeleton modulation is coordinated by multiple actin-binding proteins (ABP). Through Kyoto encyclopedia of gene and genomes database, GEPIA website, and real-time PCR data, we found that H. pylori infection significantly induced L-plastin, a key ABP, in gastric cancer cells. We further explored the regulation and function of L-plastin in H. pylori-associated gastric cancer and found that, mechanistically, H. pylori infection induced gastric cancer cells to express L-plastin via cagA-activated ERK signaling pathway to mediate SP1 binding to L-plastin promoter. Moreover, this increased L-plastin promoted gastric cancer cell proliferation and migration in vitro and facilitated the growth and metastasis of gastric cancer in vivo. Finally, we detected the expression pattern of L-plastin in gastric cancer tissues, and found that L-plastin was increased in gastric cancer tissues and that this increase of L-plastin positively correlated with cagA + H. pylori infection status. Overall, our results elucidate a novel mechanism of L-plastin expression induced by H. pylori, and a new function of L-plastin-facilitated growth and metastasis of gastric cancer, and thereby implicating L-plastin as a potential therapeutic target against gastric cancer. IMPLICATIONS: Our results elucidate a novel mechanism of L-plastin expression induced by H. pylori in gastric cancer, and a new function of L-plastin-facilitated gastric cancer growth and metastasis, implicating L-plastin as a potential therapeutic target against gastric cancer.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Helicobacter Infections/genetics , Helicobacter pylori/genetics , MAP Kinase Signaling System/genetics , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Sp1 Transcription Factor/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Humans , Male , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Microfilament Proteins/metabolism , Middle Aged , Neoplasm Metastasis , Sp1 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Transplantation, Heterologous
12.
ACS Appl Mater Interfaces ; 12(50): 55584-55595, 2020 Dec 16.
Article in English | MEDLINE | ID: mdl-33259182

ABSTRACT

Magnetic nanostructures (MNS) have a wide range of biological applications due to their biocompatibility, superparamagnetic properties, and customizable composition that includes iron oxide (Fe3O4), Zn2+, and Mn2+. However, several challenges to the biomedical usage of MNS must still be addressed, such as formulation stability, inability to encapsulate therapeutic payloads, and variable clearance rates in vivo. Here, we enhance the utility of MNS during controlled delivery applications via encapsulation within polymeric bicontinuous nanospheres (BCNs) composed of poly(ethylene glycol)-block-poly(propylene sulfide) (PEG-b-PPS) copolymers. PEG-b-PPS BCNs have demonstrated versatile encapsulation and delivery capabilities for both hydrophilic and hydrophobic payloads due to their unique and highly organized cubic phase nanoarchitecture. MNS-embedded BCNs (MBCNs) were thus coloaded with physicochemically diverse molecular payloads using the technique of flash nanoprecipitation and characterized in terms of their structure and in vivo biodistribution following intravenous administration. Retention of the internal aqueous channels and cubic architecture of MBCNs were verified using cryogenic transmission electron microscopy and small-angle X-ray scattering, respectively. MBCNs demonstrated improvement in magnetic resonance imaging (MRI) contrast enhancement (r2 relaxivity) as compared to free MNS, which in combination with scanning transmission electron microscopy and energy-dispersive X-ray spectroscopy evidenced the clustering and continued access to water of MNS following encapsulation. Furthermore, MBCNs were found to be noncytotoxic and able to deliver their hydrophilic and hydrophobic small-molecule payloads both in vitro and in vivo. Finally, the oxidation sensitivity of the hydrophobic PPS block allowed MBCNs to undergo a unique, triggerable transition in morphology into MNS-bearing micellar nanocarriers. In summary, MBCNs are an attractive platform for the delivery of molecular and nanoscale payloads for diverse on-demand and sustained drug delivery applications.


Subject(s)
Magnetite Nanoparticles/chemistry , Nanospheres/chemistry , Animals , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/toxicity , Female , Ferrosoferric Oxide/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Liver/chemistry , Liver/metabolism , MCF-7 Cells , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Nanospheres/metabolism , Nanospheres/toxicity , Oxidation-Reduction , Polyethylene Glycols/chemistry , Sulfides/chemistry , Tissue Distribution
13.
Surg Oncol ; 34: 24-30, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32891337

ABSTRACT

BACKGROUND: Surgical is the optimal therapeutic strategy for sacral tumors, and complete resection can effectively improve the recurrence and survival rates. However, the specialized anatomy, massive bleeding and adhesion to the anterior tissue, especially that caused by giant sacral tumors, makes complete resection difficult. The laparoscopic technique provides a new method to resect sacral tumors. METHODS: 34 patients with primary giant sacral tumors who underwent surgical resection were enrolled. After bilateral internal iliac artery ligation and anterior laparoscopic tumor separation, the sacral tumors were successfully resected posteriorly. The clinical, radiological and follow-up data were collected and analyzed. RESULTS: The average operative time was 276.47Ā min and that for laparoscopy was 76.24Ā min. The average intraoperative blood loss was 1757.64Ā ml. No complications associated with laparoscopic surgery, such as intestinal, urinary tract, or vascular injuries, occurred. Ten patients (29.41%) had perioperative complications, including infection, unhealed wounds, and cerebrospinal fluid leaks in 10, 5 and 2 patients, respectively. Patients with complications had significantly longer total (55.00Ā Ā±Ā 34.53 vs 25.13Ā Ā±Ā 14.60, PĀ =Ā 0.001) and postoperative (39.10Ā Ā±Ā 30.61 vs 14.83Ā Ā±Ā 10.00, PĀ =Ā 0.002) hospitalization stays than patients without complications. Postoperatively, bowel and bladder dysfunction, intestinal obstruction, pain, and perianal numbness occurred in 21, 5, 8, and 2 patients, respectively. The recurrence rate was 11.76%. CONCLUSIONS: Laparoscopically assisted sacral tumor resection is a technically feasible and effective surgical method to resect giant sacral tumors, with the advantages of reduced operative blood loss during internal iliac artery ligation and anterior tumor separation.


Subject(s)
Giant Cell Tumors/surgery , Iliac Artery/surgery , Laparoscopy/methods , Sacrum/surgery , Spinal Neoplasms/surgery , Adult , Aged , Female , Follow-Up Studies , Giant Cell Tumors/pathology , Humans , Iliac Artery/pathology , Ligation , Male , Middle Aged , Prognosis , Retrospective Studies , Sacrum/pathology , Spinal Neoplasms/pathology , Young Adult
14.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32634127

ABSTRACT

Arrestin domain containing 3 (ARRDC3) represents a newly discovered α-arrestin involved in obesity, inflammation, and cancer. Here, we demonstrate a proinflammation role of ARRDC3 in Helicobacter pylori-associated gastritis. Increased ARRDC3 was detected in gastric mucosa of patients and mice infected with H. pylori. ARRDC3 in gastric epithelial cells (GECs) was induced by H. pylori, regulated by ERK and PI3K-AKT pathways in a cagA-dependent manner. Human gastric ARRDC3 correlated with the severity of gastritis, and mouse ARRDC3 from non-BM-derived cells promoted gastric inflammation. This inflammation was characterized by the CXCR2-dependent influx of CD45+CD11b+Ly6C-Ly6G+ neutrophils, whose migration was induced via the ARRDC3-dependent production of CXCL2 by GECs. Importantly, gastric inflammation was attenuated in Arrdc3-/- mice but increased in protease-activated receptor 1-/- (Par1-/-) mice. Mechanistically, ARRDC3 in GECs directly interacted with PAR1 and negatively regulated PAR1 via ARRDC3-mediated lysosomal degradation, which abrogated the suppression of CXCL2 production and following neutrophil chemotaxis by PAR1, thereby contributing to the development of H. pylori-associated gastritis. This study identifies a regulatory network involving H. pylori, GECs, ARRDC3, PAR1, and neutrophils, which collectively exert a proinflammatory effect within the gastric microenvironment. Efforts to inhibit this ARRDC3-dependent pathway may provide valuable strategies in treating of H. pylori-associated gastritis.


Subject(s)
Arrestins/metabolism , Arrestins/physiology , Gastric Mucosa/pathology , Gastritis/pathology , Helicobacter Infections/complications , Inflammation/pathology , Receptor, PAR-1/physiology , Animals , Arrestins/genetics , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastritis/metabolism , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Inflammation/metabolism , Inflammation/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Cell Death Dis ; 11(3): 189, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32184393

ABSTRACT

Adrenomedullin (ADM) is a multifunctional peptide that is expressed by many surface epithelial cells, but its relevance to Helicobacter pylori (H. pylori)-induced gastritis is unknown. Here, we found that gastric ADM expression was elevated in gastric mucosa of H. pylori-infected patients and mice. In H. pylori-infected human gastric mucosa, ADM expression was positively correlated with the degree of gastritis; accordingly, blockade of ADM resulted in decreased inflammation within the gastric mucosa of H. pylori-infected mice. During H. pylori infection, ADM production was promoted via PI3K-AKT signaling pathway activation by gastric epithelial cells in a cagA-dependent manner, and resulted in increased inflammation within the gastric mucosa. This inflammation was characterized by the increased IFN-ƎĀ³-producing T cells, whose differentiation was induced via the phosphorylation of AKT and STAT3 by ADM derived from gastric epithelial cells. ADM also induced macrophages to produce IL-12, which promoted the IFN-ƎĀ³-producing T-cell responses, thereby contributing to the development of H. pylori-associated gastritis. Accordingly, blockade of IFN-ƎĀ³ or knockout of IFN-ƎĀ³ decreased inflammation within the gastric mucosa of H. pylori-infected mice. This study identifies a novel regulatory network involving H. pylori, gastric epithelial cells, ADM, macrophages, T cells, and IFN-ƎĀ³, which collectively exert a pro-inflammatory effect within the gastric microenvironment.


Subject(s)
Adrenomedullin/adverse effects , Gastritis/genetics , Helicobacter pylori/pathogenicity , Interferon-gamma/metabolism , T-Lymphocytes/metabolism , Vasodilator Agents/adverse effects , Animals , Gastritis/metabolism , Humans , Mice
16.
Biomater Sci ; 7(2): 657-668, 2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30601470

ABSTRACT

In this work, the hydrophobic small molecule NF-κB inhibitor celastrol was loaded into poly(ethylene glycol)-b-poly(propylene sulfide) (PEG-b-PPS) micelles. PEG-b-PPS micelles demonstrated high loading efficiency, low polydispersity, and no morphological changes upon loading with celastrol. Encapsulation of celastrol within these nanocarriers significantly reduced cytotoxicity compared to free celastrol, while simultaneously expanding the lower concentration range for effective inhibition of NF-κB signaling by nearly 50 000-fold. Furthermore, celastrol-loaded micelles successfully reduced TNF-α secretion after LPS stimulation of RAW 264.7 cells and reduced the number of neutrophils and inflammatory monocytes within atherosclerotic plaques of ldlr-/- mice. This reduction in inflammatory cells was matched by a reduction in plaque area, suggesting that celastrol-loaded nanocarriers may serve as an anti-inflammatory treatment for atherosclerosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Drug Carriers/chemistry , Plaque, Atherosclerotic/drug therapy , Polyethylene Glycols/chemistry , Sulfides/chemistry , Triterpenes/chemistry , Triterpenes/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Female , Mice , Mice, Inbred C57BL , Micelles , NF-kappa B/metabolism , Pentacyclic Triterpenes , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , RAW 264.7 Cells , Signal Transduction/drug effects , Triterpenes/pharmacology
17.
Sci Adv ; 5(4): eaau6547, 2019 04.
Article in English | MEDLINE | ID: mdl-30949574

ABSTRACT

The interaction between gastric epithelium and immune response plays key roles in H. pylori-associated pathology. We demonstrated a procolonization and proinflammation role of MMP-10 in H. pylori infection. MMP-10 is elevated in gastric mucosa and is produced by gastric epithelial cells synergistically induced by H. pylori and IL-22 via the ERK pathway. Human gastric MMP-10 was correlated with H. pylori colonization and the severity of gastritis, and mouse MMP-10 from non-BM-derived cells promoted bacteria colonization and inflammation. H. pylori colonization and inflammation were attenuated in IL-22-/-, MMP-10-/-, and IL-22-/-MMP-10-/- mice. MMP-10-associated inflammation is characterized by the influx of CD8+ T cells, whose migration is induced via MMP-10-CXCL16 axis by gastric epithelial cells. Under the influence of MMP-10, Reg3a, E-cadherin, and zonula occludens-1 proteins decrease, resulting in impaired host defense and increased H. pylori colonization. Our results suggest that MMP-10 facilitates H. pylori persistence and promotes gastritis.


Subject(s)
Gastritis/metabolism , Gastritis/microbiology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Matrix Metalloproteinase 10/metabolism , Animals , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL16/metabolism , Disease Models, Animal , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gene Expression , Helicobacter Infections/genetics , Humans , Interleukins/metabolism , Matrix Metalloproteinase 10/genetics , Mice , Mice, Knockout , Models, Biological , Interleukin-22
18.
Cell Death Dis ; 10(2): 79, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692510

ABSTRACT

Interleukin-17 receptor B (IL-17RB), a member of the IL-17 receptor family activated by IL-17B/IL-17E, has been shown to be involved in inflammatory diseases. However, the regulation and function of IL-17RB in Helicobacter pylori (H. pylori) infection, especially in the early-phase is still unknown. Here, we found that gastric IL-17RB mRNA and protein were decreased in gastric mucosa of both patients and mice infected with H. pylori. In vitro experiments show that IL-17RB expression was down regulated via PI3K/AKT pathway on gastric epithelial cells (GECs) stimulated with H. pylori in a cagA-involved manner, while in vivo studies showed that the effect was partially dependent on cagA expression. IL-17E was also decreased during the early-phase of H. pylori infection, and provision of exogenous IL-17E resulted in increased CD11b+CD11c- myeloid cells accumulation and decreased bacteria colonization within the gastric mucosa. In the early-phase of H. pylori infection, IL-17E-IL-17RB promoted gastric epithelial cell-derived CXCL1/2/5/6 to attract CD11b+CD11c- myeloid cells, and also contributed to host defense by promoting the production of antibacterial protein Reg3a. This study defines a negative regulatory network involving IL-17E, GECs, IL-17RB, CD11b+CD11c- myeloid cells, and Reg3a in the early-phase of H. pylori infection, which results in an impaired host defense within the gastric microenvironment, suggesting IL-17RB as a potential early intervening target in H. pylori infection.


Subject(s)
CD11b Antigen/immunology , CD11c Antigen/immunology , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Helicobacter pylori/isolation & purification , Myeloid Cells/immunology , Receptors, Interleukin-17/immunology , Animals , CD11 Antigens/biosynthesis , CD11 Antigens/immunology , CD11b Antigen/biosynthesis , CD11b Antigen/blood , CD11c Antigen/biosynthesis , Helicobacter Infections/blood , Helicobacter Infections/genetics , Helicobacter pylori/genetics , Humans , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, Interleukin-17/biosynthesis , Receptors, Interleukin-17/genetics
19.
Chin J Traumatol ; 11(3): 161-4, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18507946

ABSTRACT

OBJECTIVE: To study the expression regularity of vascular endothelial growth factor (VEGF) during the process of fracture healing, and the type of VEGF receptor expressed in the vascular endothelial cells of the fracture site. METHODS: The fracture model was made in the middle part of left radius in 35 rabbits. The specimens from the fracture site were harvested at 8, 24, 72 hours and 1, 3, 5, 8 weeks, and then fixed, decalcified, and sectioned frozenly to detect the expression of VEGF and its receptor at the fracture site by in situ hybridization and immunochemical assays. RESULTS: VEGF mRNA and VEGF expression was detected in many kinds of cells at the fracture site during 8 hours to 8 weeks after fracture. Flt1 receptor of VEGF was found in the vascular endothelial cells at the fracture site during 8 hours to 8 weeks after fracture, and strong expression of flk1 receptor was detected from 3 days to 3 weeks after fracture. CONCLUSIONS: The expression of VEGF and flt1 receptor appears during the whole course of fracture healing, especially from 1 to 3 weeks. Flk1 receptor is highly expressed in a definite period after fracture. VEGF is proved to be involved in the vascular reconstruction and fracture healing.


Subject(s)
Endothelial Cells/chemistry , Fracture Healing/physiology , Receptors, Vascular Endothelial Growth Factor/analysis , Vascular Endothelial Growth Factor A/analysis , Animals , Female , Immunohistochemistry , In Situ Hybridization , Male , Rabbits
20.
Cell Death Dis ; 9(10): 1034, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30305610

ABSTRACT

Mast cells are prominent components of solid tumors and exhibit distinct phenotypes in different tumor microenvironments. However, their precise mechanism of communication in gastric cancer remains largely unclear. Here, we found that patients with GC showed a significantly higher mast cell infiltration in tumors. Mast cell levels increased with tumor progression and independently predicted reduced overall survival. Tumor-derived adrenomedullin (ADM) induced mast cell degranulation via PI3K-AKT signaling pathway, which effectively promoted the proliferation and inhibited the apoptosis of GC cells in vitro and contributed to the growth and progression of GC tumors in vivo, and the effect could be reversed by blocking interleukin (IL)-17A production from these mast cells. Our results illuminate a novel protumorigenic role and associated mechanism of mast cells in GC, and also provide functional evidence for these mast cells to prevent, and to treat this immunopathogenesis feature of GC.


Subject(s)
Adrenomedullin/metabolism , Mast Cells/metabolism , Mast Cells/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Exocytosis/physiology , Female , Humans , Interleukin-17/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Stomach/pathology , Tumor Microenvironment/physiology
SELECTION OF CITATIONS
SEARCH DETAIL