Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Exp Clin Cancer Res ; 41(1): 132, 2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35392977

ABSTRACT

BACKGROUND: Interactions between tumor and microenvironment determine individual response to immunotherapy. Triple negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) have exhibited suboptimal responses to immune checkpoint inhibitors (ICIs). Aspartate ß-hydroxylase (ASPH), an oncofetal protein and tumor associated antigen (TAA), is a potential target for immunotherapy. METHODS: Subcutaneous HCC and orthotopic TNBC murine models were established in immunocompetent BALB/c mice with injection of BNL-T3 and 4 T1 cells, respectively. Immunohistochemistry, immunofluorescence, H&E, flow cytometry, ELISA and in vitro cytotoxicity assays were performed. RESULTS: The ASPH-MYC signaling cascade upregulates PD-L1 expression on breast and liver tumor cells. A bio-nanoparticle based λ phage vaccine targeting ASPH was administrated to mice harboring syngeneic HCC or TNBC tumors, either alone or in combination with PD-1 blockade. In control, autocrine chemokine ligand 13 (CXCL13)-C-X-C chemokine receptor type 5 (CXCR5) axis promoted tumor development and progression in HCC and TNBC. Interactions between PD-L1+ cancer cells and PD-1+ T cells resulted in T cell exhaustion and apoptosis, causing immune evasion of cancer cells. In contrast, combination therapy (Vaccine+PD-1 inhibitor) significantly suppressed primary hepatic or mammary tumor growth (with distant pulmonary metastases in TNBC). Adaptive immune responses were attributed to expansion of activated CD4+ T helper type 1 (Th1)/CD8+ cytotoxic T cells (CTLs) that displayed enhanced effector functions, and maturation of plasma cells that secreted high titers of ASPH-specific antibody. Combination therapy significantly reduced tumor infiltration of immunosuppressive CD4+/CD25+/FOXP3+ Tregs. When the PD-1/PD-L1 signal was inhibited, CXCL13 produced by ASPH+ cancer cells recruited CXCR5+/CD8+ T lymphocytes to tertiary lymphoid structures (TLSs), comprising effector and memory CTLs, T follicular helper cells, B cell germinal center, and follicular dendritic cells. TLSs facilitate activation and maturation of DCs and actively recruit immune subsets to tumor microenvironment. These CTLs secreted CXCL13 to recruit more CXCR5+ immune cells and to lyse CXCR5+ cancer cells. Upon combination treatment, formation of TLSs predicts sensitivity to ICI blockade. Combination therapy substantially prolonged overall survival of mice with HCC or TNBC. CONCLUSIONS: Synergistic antitumor efficacy attributable to a λ phage vaccine specifically targeting ASPH, an ideal TAA, combined with ICIs, inhibits tumor growth and progression of TNBC and HCC.


Subject(s)
Cancer Vaccines , Carcinoma, Hepatocellular , Immunotherapy , Liver Neoplasms , Triple Negative Breast Neoplasms , Animals , B7-H1 Antigen , Cancer Vaccines/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Humans , Immune Checkpoint Inhibitors , Immunity , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Mice , Nanoparticles , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment
2.
Am J Cancer Res ; 11(9): 4141-4174, 2021.
Article in English | MEDLINE | ID: mdl-34659881

ABSTRACT

Triple negative breast cancer (TNBC) is more aggressive and has a poorer prognosis than other sub-types of breast tumors. This study elucidates how aspartate beta-hydroxylase (ASPH) network promotes drug resistance, and immunotherapy targeting ASPH may improve the efficacy of Doxorubicin (DOX) therapy. An orthotopic model of breast cancer generated by 4T1 cells in immunocompetent mice was used to explore efficacy of immunotherapy in combination with DOX chemotherapy. We evaluated mRNA and protein expression in cultured tumor cells and tissue, as well as assessed cell proliferation, apoptosis, soluble factors/cytokine production, immune cell population diversity and function. We observed that ASPH expression enables TNBC cells to exhibit primary resistance to DOX induced single-/double-strand breaks (SSB/DSB) and enhanced proliferation and survival. Specific bio-nanoparticle based therapeutic vaccine (BNP-TV) promoted ASPH uptake by and maturation of DCs. This BNP-TV combined with DOX induces immunogenic cell death (ICD) in orthotopic xenograft tumors and significantly suppressed primary mammary tumor growth and distant multi-organ metastases. Immunogenic cell death induced by BNP-TV targeting ASPH combined with DOX provides opportunities to treat a highly resistant and metastatic form of breast cancer.

3.
Cells ; 10(2)2021 01 20.
Article in English | MEDLINE | ID: mdl-33498326

ABSTRACT

A recently discovered human glycoprotein, chitinase 3-like 1 (Chi3L1), may play a role in inflammation, tissue remodeling, and visceral fat accumulation. We hypothesize that Chi3L1 gene expression is important in the development of hepatic insulin resistance characterized by the generation of pAKT, pGSK, and pERK in wild type and Chi3L1 knockout (KO) murine liver following insulin stimulation. The Chi3L1 gene and protein expression was evaluated by Real Time PCR and ELISA; lipid accumulation in hepatocytes was also assessed. To alter Chi3L1 function, three different anti-Chi3L1 monoclonal antibodies (mAbs) were administered in vivo and effects on the insulin signaling cascade and hepatic lipid deposition were determined. Transmission of the hepatic insulin signal was substantially improved following KO of the CHi3L1 gene and there was reduced lipid deposition produced by a HFD. The HFD-fed mice exhibited increased Chi3L1 expression in the liver and there was impaired insulin signal transduction. All three anti-Chi3L1 mAbs partially restored hepatic insulin sensitivity which was associated with reduced lipid accumulation in hepatocytes as well. A KO of the Chi3L1 gene reduced lipid accumulation and improved insulin signaling. Therefore, Chi3L1 gene upregulation may be an important factor in the generation of NAFLD/NASH phenotype.


Subject(s)
Chitinase-3-Like Protein 1/metabolism , Insulin Resistance , Liver/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Biopsy , Chitinase-3-Like Protein 1/genetics , Diet, High-Fat , Feeding Behavior , Insulin/metabolism , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction
4.
Zhongguo Dang Dai Er Ke Za Zhi ; 22(9): 942-947, 2020 Sep.
Article in Chinese | MEDLINE | ID: mdl-32933623

ABSTRACT

OBJECTIVE: To investigate the clinical features and outcome of neonatal acute respiratory distress syndrome (ARDS) in southwest Hubei, China. METHODS: According to the Montreux definition of neonatal ARDS, a retrospective clinical epidemiological investigation was performed on the medical data of neonates with ARDS who were admitted to Department of Neonatology/Pediatrics in 17 level 2 or level 3 hospitals in southwest Hubei from January to December, 2017. RESULTS: A total of 7 150 neonates were admitted to the 17 hospitals in southwest Hubei during 2017 and 66 (0.92%) were diagnosed with ARDS. Among the 66 neonates with ARDS, 23 (35%) had mild ARDS, 28 (42%) had moderate ARDS, and 15 (23%) had severe ARDS. The main primary diseases for neonatal ARDS were perinatal asphyxia in 23 neonates (35%), pneumonia in 18 neonates (27%), sepsis in 12 neonates (18%), and meconium aspiration syndrome in 10 neonates (15%). Among the 66 neonates with ARDS, 10 neonates (15%) were born to the mothers with an age of ≥35 years, 30 neonates (45%) suffered from intrauterine distress, 32 neonates (49%) had a 1-minute Apgar score of 0 to 7 points, 24 neonates (36%) had abnormal fetal heart monitoring results, and 21 neonates (32%) experienced meconium staining of amniotic fluid. Intraventricular hemorrhage was the most common comorbidity (12 neonates), followed by neonatal shock (9 neonates) and patent ductus arteriosus (8 neonates). All 66 neonates with ARDS were treated with mechanical ventilation in addition to the treatment for primary diseases. Among the 66 neonates with ARDS, 10 died, with a mortality rate of 15% (10/66), and 56 neonates were improved or cured, with a survival rate of 85% (56/66). CONCLUSIONS: Neonatal ARDS in southwest Hubei is mostly mild or moderate. Perinatal asphyxia and infection may be the main causes of neonatal ARDS in this area. Intraventricular hemorrhage is the most common comorbidity. Neonates with ARDS tend to have a high survival rate after multimodality treatment.


Subject(s)
Respiratory Distress Syndrome, Newborn , China , Female , Humans , Infant, Newborn , Meconium Aspiration Syndrome , Pregnancy , Retrospective Studies
5.
Cancer Lett ; 481: 63-75, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32145343

ABSTRACT

To demonstrate multifaceted contribution of aspartate ß-hydroxylase (ASPH) to pancreatic ductal adenocarcinoma (PDAC) pathogenesis, in vitro metastasis assay and patient derived xenograft (PDX) murine models were established. ASPH propagates aggressive phenotypes characterized by enhanced epithelial-mesenchymal transition (EMT), 2-D/3-D invasion, extracellular matrix (ECM) degradation/remodeling, angiogenesis, stemness, transendothelial migration and metastatic colonization/outgrowth at distant sites. Mechanistically, ASPH activates Notch cascade through direct physical interactions with Notch1/JAGs and ADAMs. The ASPH-Notch axis enables prometastatic secretome trafficking via exosomes, subsequently initiates MMPs mediated ECM degradation/remodeling as an effector for invasiveness. Consequently, ASPH fosters primary tumor development and pulmonary metastasis in PDX models, which was blocked by a newly developed small molecule inhibitor (SMI) specifically against ASPH's ß-hydroxylase activity. Clinically, ASPH is silenced in normal pancreas, progressively upregulated from pre-malignant lesions to invasive/advanced stage PDAC. Relatively high levels of ASPH-Notch network components independently/jointly predict curtailed overall survival (OS) in PDAC patients (log-rank test, Ps < 0.001; Cox proportional hazards regression, P < 0.001). Therefore, ASPH-Notch axis is essential for propagating multiple-steps of metastasis and predicts prognosis of PDAC patients. A specific SMI targeting ASPH offers a novel therapeutic approach to substantially retard PDAC development/progression.


Subject(s)
Exosomes/pathology , Lung Neoplasms/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Neoplastic/physiology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Prognosis , Signal Transduction/physiology , Xenograft Model Antitumor Assays/methods , Pancreatic Neoplasms
6.
Cancer Lett ; 449: 87-98, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30768955

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy with very limited treatment options. Antibody drug conjugates (ADCs) are promising cytotoxic agents capable of highly selective delivery. Aspartate-ß-hydroxylase (ASPH) is a type II transmembrane protein highly expressed in PDACs (97.1%) but not normal pancreas. We investigated anti-tumor effects of an ADC guided by a human monoclonal antibody (SNS-622) against ASPH in human PDAC cell lines and derived subcutaneous (s.c.) xenograft as well as a patient-derived xenograft (PDX) murine model with spontaneous pulmonary metastasis. The cytotoxic effects exhibited by several candidate payloads linked to SNS-622 antibody targeting ASPH+ PDACs were analyzed. After i.v. administration of SNS-622-emtansine (DM1) ADC, the primary PDAC tumor growth and progression (number and size of pulmonary metastases) were determined. The PDAC cell lines, s.c. and PDX tumors treated with ADC were tested for cell proliferation, cytotoxicity and apoptosis by MTS and immunohistochemistry (IHC) assays. SNS-622-DM1 construct has demonstrated optimal anti-tumor effects in vitro. In the PDX model of human PDAC, SNS-622-DM1 ADC exerted substantially inhibitory effects on tumor growth and pulmonary metastasis through attenuating proliferation and promoting apoptosis.


Subject(s)
Calcium-Binding Proteins/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Immunoconjugates/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Membrane Proteins/antagonists & inhibitors , Mixed Function Oxygenases/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Administration, Intravenous , Animals , Carcinoma, Pancreatic Ductal/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/pharmacology , Lung Neoplasms/enzymology , Mice , Pancreatic Neoplasms/enzymology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
7.
Zhongguo Dang Dai Er Ke Za Zhi ; 21(1): 6-10, 2019 Jan.
Article in Chinese | MEDLINE | ID: mdl-30675856

ABSTRACT

OBJECTIVE: To investigate the incidence of neonatal asphyxia and possible contributing factors for the development of severe asphyxia in Hubei Enshi Tujia and Miao Autonomous Prefecture, China. METHODS: A total of 16 hospitals in Hubei Enshi Tujia and Miao Autonomous Prefecture were selected as research centers. A retrospective analysis was performed for the clinical data of 22 294 live births in these 16 hospitals from January to December, 2016 to investigate the incidence rate of neonatal asphyxia and possible contributing factors for the development of severe asphyxia. RESULTS: Of the 22 294 neonates born alive, 733 (3.29%) were diagnosed with neonatal asphyxia, among whom 627 had mild asphyxia and 106 had severe asphyxia. The neonates with low maternal education level, maternal anemia during pregnancy, chorioamnionitis, abnormal amniotic fluid, abnormal umbilical cord, placenta previa, placental abruption, Tujia Minority, preterm birth, and low birth weight had a higher incidence of severe asphyxia (P<0.05). CONCLUSIONS: The incidence rate of neonatal asphyxia in Hubei Enshi Tujia and Miao Autonomous Prefecture is higher. Low maternal education level, maternal anemia during pregnancy, chorioamnionitis, abnormal amniotic fluid, abnormal umbilical cord, placenta previa, placental abruption, Tujia Minority, preterm birth, and low birth weight may be related to the development of severe neonatal asphyxia.


Subject(s)
Asphyxia Neonatorum , Asphyxia Neonatorum/epidemiology , China , Humans , Incidence , Infant, Newborn , Retrospective Studies
8.
J Hematol Oncol ; 12(1): 144, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31888763

ABSTRACT

BACKGROUND: Signaling pathways critical for embryonic development re-emerge in adult pancreas during tumorigenesis. Aspartate ß-hydroxylase (ASPH) drives embryonic cell motility/invasion in pancreatic development/differentiation. We explored if dysregulated ASPH is critically involved in pancreatic cancer pathogenesis. METHODS: To demonstrate if/how ASPH mediates malignant phenotypes, proliferation, migration, 2-D/3-D invasion, pancreatosphere formation, immunofluorescence, Western blot, co-immunoprecipitation, invadopodia formation/maturation/function, qRT-PCR, immunohistochemistry (IHC), and self-developed in vitro metastasis assays were performed. Patient-derived xenograft (PDX) models of human pancreatic ductal adenocarcinoma (PDAC) were established to illustrate in vivo antitumor effects of the third-generation small molecule inhibitor specifically against ASPH's ß-hydroxylase activity. Prognostic values of ASPH network components were evaluated with Kaplan-Meier plots, log-rank tests, and Cox proportional hazards regression models. RESULTS: ASPH renders pancreatic cancer cells more aggressive phenotypes characterized by epithelial-mesenchymal transition (EMT), 2-D/3-D invasion, invadopodia formation/function as demonstrated by extracellular matrix (ECM) degradation, stemness (cancer stem cell marker upregulation and pancreatosphere formation), transendothelial migration (mimicking intravasation/extravasation), and sphere formation (mimicking metastatic colonization/outgrowth at distant sites). Mechanistically, ASPH activates SRC cascade through direct physical interaction with ADAM12/ADAM15 independent of FAK. The ASPH-SRC axis enables invadopodia construction and initiates MMP-mediated ECM degradation/remodeling as executors for invasiveness. Pharmacologic inhibition of invadopodia attenuates in vitro metastasis. ASPH fosters primary tumor development and pulmonary metastasis in PDX models of PDAC, which is blocked by a leading compound specifically against ASPH enzymatic activity. ASPH is silenced in normal pancreas, progressively upregulated from pre-malignant lesions to invasive/advanced stages of PDAC. Expression profiling of ASPH-SRC network components independently/jointly predicts clinical outcome of PDAC patients. Compared to a negative-low level, a moderate-very high level of ASPH, ADAM12, activated SRC, and MMPs correlated with curtailed overall survival (OS) of pancreatic cancer patients (log-rank test, ps < 0.001). The more unfavorable molecules patients carry, the more deleterious prognosis is destinated. Patients with 0-2 (n = 4), 3-5 (n = 8), 6-8 (n = 24), and 9-12 (n = 73) unfavorable expression scores of the 5 molecules had median survival time of 55.4, 15.9, 9.7, and 5.0 months, respectively (p < 0.001). CONCLUSION: Targeting the ASPH-SRC axis, which is essential for propagating multi-step PDAC metastasis, may specifically/substantially retard development/progression and thus improve prognosis of PDAC.


Subject(s)
Biomarkers, Tumor/metabolism , Calcium-Binding Proteins/metabolism , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/secondary , Membrane Proteins/metabolism , Mixed Function Oxygenases/metabolism , Muscle Proteins/metabolism , Pancreatic Neoplasms/pathology , src-Family Kinases/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Calcium-Binding Proteins/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mixed Function Oxygenases/genetics , Muscle Proteins/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , src-Family Kinases/genetics
9.
Carbohydr Polym ; 196: 225-232, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29891291

ABSTRACT

Inspired by the supramolecular structure of cellulose, cellulose-gelatin supramolecular hydrogels with high strength and pH-sensitivity were constructed in a basic-based solvent system, ethylene diamine/potassium thiocyanate (EDA/KSCN) with the aid of cyclic freezing-thawing. The investigation on the characteristics of supramolecular hydrogels revealed that repeated freezing-thawing cycles played an important role in the formation of the physical cross-linked supramolecular network structure between cellulose and gelatin. The mechanical properties of supramolecular hydrogels were much higher than pure cellulose and gelatin hydrogel, and the compressive strength was 9.6 times higher than that of pure gelatin hydrogel. The synergistic effect between hydrogen-bonding interaction and the reinforcement of regenerated cellulose nanofibrils (CNF) contributed to the superior mechanical performance. Furthermore, the swelling kinetics tests showed that the supramolecular hydrogels exhibited excellent pH-responsibility, indicating potential applications in biomedical fields. Thus, a straightforward route to construct natural polymer-based hydrogels with supramolecular structure through physical crosslinking strategy without employing hazardous crosslinking agents was developed, paving the way for the design of new types of hydrogels.

10.
Minerva Gastroenterol Dietol ; 64(3): 310-321, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29600697

ABSTRACT

Over the past 30 years, multidrug regimens consisting of a proton pump inhibitor (PPI) and two or three antibiotics have been used in treating Helicobacter pylori (H. pylori) infection. In clinical practice, the optimal regimen to cure H. pylori infection should be decided regionally. Considering the first treatment, the Maastricht V/Florence Consensus Report and the American College of Gastroenterology Clinical Management Guideline highlight that in countries with low clarithromycin resistance rates (<15%), an empiric clarithromycin-based regimen can be used. In countries with high clarithromycin resistance rates or, in the American Guideline, with a previous exposure to clarithromycin, a bismuth-containing quadruple therapy (with metronidazole and tetracycline) is the first choice. In case of persistent infection, after a previous clarithromycin-containing regimen, this drug should be avoided in second line therapy. Options after initial eradication failure include tailored therapy (choosing antibiotic combinations based on antibiotic susceptibility testing), empiric bismuth-containing quadruple therapy or triple levofloxacin-based therapy. Encouraging data are reported, both for the first-line and for rescue treatments, with the use of a formulation of bismuth subcitrate potassium, metronidazole, and tetracycline contained in a single capsule, together with a PPI. Rifabutin- and furazolidone-based regimens should also be considered in rescue regimens. Vonoprazan, a new type of potassium-competitive acid blocker that produces more potent acid inhibition than PPIs, provides improved H. pylori eradication rates in combination with antibiotics. In this review, the authors provide an overview on the current knowledge on the treatment of H. pylori infection, with focus on therapeutic challenges in this field.


Subject(s)
Helicobacter Infections/drug therapy , Helicobacter pylori , Anti-Bacterial Agents/therapeutic use , Clarithromycin/therapeutic use , Clinical Protocols , Drug Resistance, Bacterial , Humans , Pyrroles/therapeutic use , Sulfonamides/therapeutic use
12.
Oncotarget ; 7(43): 69136-69148, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27655701

ABSTRACT

OBJECTIVE: Chronic Helicobacter pylori (H. pylori) infection promotes non-cardia gastric cancer. Some mouse models suggest that bone marrow derived cells (BMDC) contribute to Helicobacter-associated gastric carcinogenesis. We determined whether this increased susceptibility to Helicobacter-induced gastric carcinogenesis of p27-deficient mice is dependent upon their p27-null BMDC or their p27-null gastric epithelial cells. DESIGN: Female mice (recipients) were irradiated and transplanted with BMDC from male donors. Wild type (WT) mice in group 1 (control) received BMDC from male GFP-transgenic mice. Female WT and p27 KO mice were engrafted with male p27KO mice BMDC (Group 2) or GFP-transgenic WT BMDC (Group 3). Recipients were infected with H. pylori SS1 for one year. RESULTS: Mice lacking p27 in either the BM pool or gastric epithelium developed significantly more advanced gastric pathology, including high-grade dysplasia. Co-staining of donor BMDC in dysplastic gastric glands was confirmed by immunofluorescence. Gastric expression of IL-1 beta protein was reduced in groups 2 and 3 (p < 0.05 vs control) whereas expression of IFN-γ and chemokines MIP-1 beta, MIG, IP-10 and RANTES in group 2 were significantly higher than group 3. CONCLUSIONS: Both bone marrow-derived and gastric epithelial cells contribute to the increased gastric cancer susceptibility of p27-deficient H. pylori-infected mice.


Subject(s)
Bone Marrow Cells/metabolism , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Helicobacter Infections/metabolism , Stomach Neoplasms/metabolism , Animals , Bone Marrow Cells/microbiology , Bone Marrow Transplantation/methods , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mosaicism , Stomach Neoplasms/microbiology
13.
Int J Clin Exp Pathol ; 8(9): 10444-51, 2015.
Article in English | MEDLINE | ID: mdl-26617753

ABSTRACT

AIM: To investigate the effect of mitogen-activated protein kinase 7 (MAPK7) in ovarian cancer metastasis and to explore its potential mechanism. METHODS: pcDNA-MAPK7 and siRNA-MAPK7 vectors were transfected into the human ovarian cell line OVCAR-3 based on gene silencing and overexpression methods. Effects of MAPK7 overexpression and silencing on OVCAR-3 cells proliferation, cell invasion, and migration were analyzed using the MTT (3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl tetrazolium bromide) assay, Matrigel methods, and Markered methods respectively. In addition, effect of MAPK7 expression on extracellular matrix (ECM) associated protein was detected using Western blot. RESULTS: Compared with the controls, MAPK7 was up-regulated when cells were transfected with pcDNA-MAPK7 plasma, as well as MAPK7 was sliced when cells were transfected with siRNA-MAPK7 plasma (P<0.05). Besides, biological function analysis performed that overexpression of MAPK7 significantly increased OVCAR-3 cell proliferation, invasion, and migration (P<0.05), while these effects were inhibited by MAPK7 silencing (P<0.05). Additionally, MAPK7 overexpression increased type II collagen expression (P<0.05). However, there was no significant difference between MAPK7 expression and type I collagen expression (P>0.05). CONCLUSION: Our data implied the up-regulated MAPK7 might contribute to ovarian cancer metastasis through up-regulating type II collagen expression and then were involved in cell biological processes such as cell proliferation, invasion, and migration. MAPK7 may be a potential therapeutic target in the clinical treatment for ovarian cancer.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Neoplasm Invasiveness/pathology , Ovarian Neoplasms/pathology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/physiology , Female , Gene Knockdown Techniques , Humans , Ovarian Neoplasms/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Up-Regulation
14.
Cancer Lett ; 359(2): 345-51, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25853150

ABSTRACT

H. pylori infection causes gastritis, peptic ulcers and gastric cancer. Eradicating H. pylori prevents ulcers, but to what extent this prevents cancer remains unknown, especially if given after intestinal metaplasia has developed. H. pylori infected wild-type (WT) mice do not develop cancer, but mice lacking the tumor suppressor p27 do so, thus providing an experimental model of H. pylori-induced cancer. We infected p27-deficient mice with H. pylori strain SS1 at 6-8 weeks of age. Persistently H. pylori-infected WT C57BL/6 mice served as controls. Mice in the eradication arms received antimicrobial therapy (omeprazole, metronidazole and clarithromycin) either "early" (at 15 weeks post infection, WPI) or "late" at 45 WPI. At 70 WPI, mice were euthanized for H. pylori determination, histopathology and cytokine/chemokine expression. Persistently infected mice developed premalignant lesions including high-grade dysplasia, whereas those given antibiotics did not. Histologic activity scores in the eradication groups were similar to each other, and were significantly decreased compared with controls for inflammation, epithelial defects, hyperplasia, metaplasia, atrophy and dysplasia. IP-10 and MIG levels in groups that received antibiotics were significantly lower than controls. There were no significant differences in expression of IFN-γ, TNF-α, IL-1ß, RANTES, MCP-1, MIP-1α or MIP-1ß among the three groups. Thus, H. pylori eradication given either early or late after infection significantly attenuated gastric inflammation, gastric atrophy, hyperplasia, and dysplasia in the p27-deficient mice model of H. pylori-induced gastric cancer, irrespective of the timing of antibiotic administration. This was associated with reduced expression of IP-10 and MIG.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Helicobacter Infections/complications , Stomach Neoplasms/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Chemokine CXCL10/blood , Chemokine CXCL10/genetics , Chemokine CXCL9/blood , Chemokine CXCL9/genetics , Clarithromycin/pharmacology , Clarithromycin/therapeutic use , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gene Expression , Helicobacter Infections/drug therapy , Helicobacter pylori/immunology , Humans , Metronidazole/pharmacology , Metronidazole/therapeutic use , Mice, Inbred C57BL , Omeprazole/pharmacology , Omeprazole/therapeutic use , Precancerous Conditions/drug therapy , Precancerous Conditions/microbiology
15.
Cancer Lett ; 355(1): 106-12, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25218349

ABSTRACT

H. pylori infection causes gastritis, peptic ulcers and gastric cancer. Eradicating H. pylori prevents ulcers, but to what extent this prevents cancer remains unknown, especially if given after intestinal metaplasia has developed. H. pylori infected wild-type (WT) mice do not develop cancer, but mice lacking the tumor suppressor p27 do so, thus providing an experimental model of H. pylori-induced cancer. We infected p27-deficient mice with H. pylori strain SS1 at 6-8 weeks of age. Persistently H. pylori-infected WT C57BL/6 mice served as controls. Mice in the eradication arms received antimicrobial therapy (omeprazole, metronidazole and clarithromycin) either "early" (at 15 weeks post infection, WPI) or "late" at 45 WPI. At 70 WPI, mice were euthanized for H. pylori determination, histopathology and cytokine/chemokine expression. Persistently infected mice developed premalignant lesions including high-grade dysplasia, whereas those given antibiotics did not. Histologic activity scores in the eradication groups were similar to each other, and were significantly decreased compared with controls for inflammation, epithelial defects, hyperplasia, metaplasia, atrophy and dysplasia. IP-10 and MIG levels in groups that received antibiotics were significantly lower than controls. There were no significant differences in expression of IFN-γ, TNF-α, IL-1ß, RANTES, MCP-1, MIP-1α or MIP-1ß among the three groups. Thus, H. pylori eradication given either early or late after infection significantly attenuated gastric inflammation, gastric atrophy, hyperplasia, and dysplasia in the p27-deficient mice model of H. pylori-induced gastric cancer, irrespective of the timing of antibiotic administration. This was associated with reduced expression of IP-10 and MIG.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Gastritis/prevention & control , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Stomach Neoplasms/prevention & control , Stomach/drug effects , Animals , Atrophy , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytokines/metabolism , Disease Models, Animal , Drug Administration Schedule , Drug Therapy, Combination , Gastric Mucosa/metabolism , Gastritis/genetics , Gastritis/metabolism , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Hyperplasia , Inflammation Mediators/metabolism , Metaplasia , Mice, Inbred C57BL , Mice, Knockout , Proton Pump Inhibitors/administration & dosage , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Time Factors
16.
PLoS One ; 9(4): e94974, 2014.
Article in English | MEDLINE | ID: mdl-24740005

ABSTRACT

H. pylori persists in the human stomach over decades and promotes several adverse clinical sequelae including gastritis, peptic ulcers and gastric cancer that are linked to the induction and subsequent evasion of chronic gastric inflammation. Emerging evidence indicates that H. pylori infection may also protect against asthma and some other immune-mediated conditions through regulatory T cell effects outside the stomach. To characterize the complexity of the CD4+ T cell response generated during H. pylori infection, computational methods were previously used to generate a panel of 90 predicted epitopes conserved among H. pylori genomes that broadly cover HLA Class II diversity for maximum population coverage. Here, these sequences were tested individually for their ability to induce in vitro responses in peripheral blood mononuclear cells by interferon-γ ELISpot assay. The average number of spot-forming cells/million PBMCs was significantly elevated in H. pylori-infected subjects over uninfected persons. Ten of the 90 peptides stimulated IFN-γ secretion in the H. pylori-infected group only, whereas two out of the 90 peptides elicited a detectable IFN-γ response in the H. pylori-uninfected subjects but no response in the H. pylori-infected group. Cytokine ELISA measurements performed using in vitro PBMC culture supernatants demonstrated significantly higher levels of TNF-α, IL-2, IL-4, IL-6, IL-10, and TGF-ß1 in the H. pylori-infected subjects, whereas IL-17A expression was not related to the subjects H. pylori-infection status. Our results indicate that the human T cell responses to these 90 peptides are generally increased in actively H. pylori-infected, compared with H. pylori-naïve, subjects. This information will improve understanding of the complex immune response to H. pylori, aiding rational epitope-driven vaccine design as well as helping identify other H. pylori epitopes with potentially immunoregulatory effects.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA-D Antigens/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Amino Acid Sequence , Computational Biology/methods , Cytokines/immunology , Cytokines/metabolism , Enzyme-Linked Immunospot Assay , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , Female , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/physiology , Host-Pathogen Interactions/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Molecular Sequence Data , Peptides/genetics , Peptides/immunology
17.
Methods Mol Biol ; 921: 89-98, 2012.
Article in English | MEDLINE | ID: mdl-23015495

ABSTRACT

Animal models are essential for in vivo analysis of Helicobacter-related diseases. Transgenic mice and Mongolian gerbil models have been the corner stone of present research focusing on both bacterial virulence factors and host response to infection. Establishing a reproducible rodent model of persistent Helicobacter pylori infection that resembles the H. pylori-associated gastritis observed in humans was a considerable challenge until Lee et al. (Gastroenterology 112:1386-1397, 1997) successfully adapted a clinical Cag A- and Vac A-expressing strain for the mouse stomach. This so-called SS1 (Sydney) strain has since been extensively used for H. pylori research; other rodent-adapted Helicobacter strains have subsequently been developed and utilized in wild-type and genetically engineered rodent models. These bacteria include both H. pylori and the larger but related species H. felis (originally isolated from cats). In this chapter we focus mainly on these two Helicobacter strains and review the rodent models that have been employed to investigate how Helicobacter species induce gastric inflammation and disease.


Subject(s)
Disease Models, Animal , Helicobacter Infections , Inflammation , Animals , Helicobacter Infections/complications , Helicobacter Infections/genetics , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/microbiology , Species Specificity , Stomach Neoplasms/chemically induced , Stomach Neoplasms/microbiology , Stomach Neoplasms/prevention & control
18.
J Immunol Methods ; 384(1-2): 157-63, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-22814402

ABSTRACT

Helicobacter pylori infection is associated with severe chronic inflammation, yet the host immune response is rarely able to clear the bacterium. Thymus derived lymphocyte populations such as T helper 1, T helper 17, and T regulatory cells are known to play important roles in the chronicity of H. pylori infection as well as contributing to ongoing gastric pathology. It is yet to be established how these immune cell populations interact in the gastric environment during H. pylori infection. Mouse models of infection offer an opportunity to investigate these interactions in detail. Flow cytometric analysis provides excellent lymphocyte characterization due to its high specificity, sensitivity and potential to perform multiple simultaneous measurements. However, this requires a viable enriched single cell suspension after adequate tissue dissociation, which poses a challenge due to the heterogeneity of gastric tissue. We have evaluated several isolation techniques and have optimized a protocol to isolate and enrich lymphocytes from the H. pylori-infected murine stomach. EDTA/DTT followed by Collagenase IV digestion successfully dissociates an average of 1 × 107 cells per mouse. Further enrichment using Lympholyte M gradient yields on average 4 × 106 CD45+ lymphocytes per stomach. Following isolation we compared lymphocyte stimulation by CD3/CD28, phorbol 12-myristate 13-acetate (PMA) and ionomycin or H. pylori lysate and determined that CD3/CD28 effectively induces stimulation of IFNγ and IL 17A, but impairs Foxp3 expression. Using an optimized protocol we observed a 2-fold increase of CD8+ IFNγ-expressing lymphocytes localized specifically to the gastric compartment during H. pylori infection. The mechanisms of H. pylori immunopathogenesis are still considered enigmatic, therefore this optimized protocol can help delineate further novel immune cell targets that mediate H. pylori-induced pathology and identify the correlates of immunity for vaccine development.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Immunophenotyping/methods , Animals , Antibodies/immunology , Antibodies/pharmacology , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Separation , Cell Survival/immunology , Cells, Cultured , Collagenases/metabolism , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Ionomycin/pharmacology , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred C57BL , Stomach/immunology , Stomach/microbiology , Stomach/pathology , Tetradecanoylphorbol Acetate/pharmacology
19.
Hum Vaccin ; 7(11): 1153-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22048119

ABSTRACT

Helicobacter pylori was appreciated as the major cause of peptic ulcers about 30 years ago and the most significant etiological agent in gastric cancer in the mid-1990s. Since that time, progress in the development of a preventive or therapeutic H. pylori vaccine has been relatively slow. The impediments to rapid advances in the field include a luke-warm enthusiasm among clinicians, research scientists, and public health authorities concerning the need for a vaccine, rudimentary understanding of the correlates of gastric immunity to H. pylori and of gastric mucosal immunology in general, the geographical heterogeneity of the H. pylori genome and insufficient pharmaceutical industry support. Recent enhancements in our understanding of the gastric immune response together with advances in H. pylori genomics now provide the potential to accelerate progress in H. pylori vaccine development. Whether an H. pylori vaccine becomes a reality will likely depend upon our ability to appropriately target the populations at highest risk of the adverse sequelae of infection.


Subject(s)
Bacterial Vaccines/therapeutic use , Gastric Mucosa/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Animals , Bacterial Vaccines/genetics , Gastric Mucosa/virology , Helicobacter Infections/immunology , Helicobacter pylori/classification , Helicobacter pylori/genetics , Humans , Mice , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use
20.
Vaccine ; 29(11): 2085-91, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21236233

ABSTRACT

Helicobacter pylori is the leading cause of gastritis, peptic ulcer disease and gastric adenocarcinoma and lymphoma in humans. Due to the decreasing efficacy of anti-H. pylori antibiotic therapy in clinical practice, there is renewed interest in the development of anti-H. pylori vaccines. In this study an in silico-based approach was utilized to develop a multi-epitope DNA-prime/peptide-boost immunization strategy using informatics tools. The efficacy of this construct was then assessed as a therapeutic vaccine in a mouse model of gastric cancer induced by chronic H. pylori infection. The multi-epitope vaccine administered intranasally induced a broad immune response as determined by interferon-gamma production in ELISpot assays. This was associated with a significant reduction in H. pylori colonization compared with mice immunized with the same vaccine intramuscularly, given an empty plasmid, or given a whole H. pylori lysate intranasally as the immunogen. Total scores of gastric histological changes were not significantly different among the 4 experimental groups. These results suggest that further development of an epitope-based mucosal vaccine may be beneficial in eradicating H. pylori and reducing the burden of the associated gastric diseases in humans.


Subject(s)
Bacterial Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Helicobacter Infections/therapy , Stomach Neoplasms/therapy , Amino Acid Sequence , Animals , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Female , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Immunity, Cellular , Immunization, Secondary , Interferon-gamma/immunology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Stomach/pathology , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology , Vaccines, DNA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL