Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Metabolites ; 14(6)2024 May 29.
Article in English | MEDLINE | ID: mdl-38921447

ABSTRACT

For either healthy or diseased organisms, lipids are key components for cellular membranes; they play important roles in numerous cellular processes including cell growth, proliferation, differentiation, energy storage and signaling. Exercise and disease development are examples of cellular environment alterations which produce changes in these networks. There are indications that alterations in lipid metabolism contribute to the development and progression of a variety of cancers. Measuring such alterations and understanding the pathways involved is critical to fully understand cellular metabolism. The demands for this information have led to the emergence of lipidomics, which enables the large-scale study of lipids using mass spectrometry (MS) techniques. Mass spectrometry has been widely used in lipidomics and allows us to analyze detailed lipid profiles of cancers. In this article, we discuss emerging strategies for lipidomics by mass spectrometry; targeted, as opposed to global, lipid analysis provides an exciting new alternative method. Additionally, we provide an introduction to lipidomics, lipid categories and their major biological functions, along with lipidomics studies by mass spectrometry in cancer samples. Further, we summarize the importance of lipid metabolism in oncology and tumor microenvironment, some of the challenges for lipodomics, and the potential for targeted approaches for screening pharmaceutical candidates to improve the therapeutic efficacy of treatment in cancer patients.

2.
Metabolites ; 14(6)2024 May 29.
Article in English | MEDLINE | ID: mdl-38921449

ABSTRACT

Acute inflammation is the body's first defense in response to pathogens or injury that is partially governed by a novel genus of endogenous lipid mediators that orchestrate the resolution of inflammation, coined specialized pro-resolving mediators (SPMs). SPMs, derived from omega-3-polyunstaturated fatty acids (PUFAs), include the eicosapentaenoic acid-derived and docosahexaenoic acid-derived Resolvins, Protectins, and Maresins. Herein, we review their biosynthesis, structural characteristics, and therapeutic effectiveness in various diseases such as ischemia, viral infections, periodontitis, neuroinflammatory diseases, cystic fibrosis, lung inflammation, herpes virus, and cancer, especially focusing on therapeutic effectiveness in respiratory inflammation and ischemia-related injuries. Resolvins are sub-nanomolar potent agonists that accelerate the resolution of inflammation by reducing excessive neutrophil infiltration, stimulating macrophage functions including phagocytosis, efferocytosis, and tissue repair. In addition to regulating neutrophils and macrophages, Resolvins control dendritic cell migration and T cell responses, and they also reduce the pro-inflammatory cytokines, proliferation, and metastasis of cancer cells. Importantly, several lines of evidence have demonstrated that Resolvins reduce tumor progression in melanoma, oral squamous cell carcinoma, lung cancer, and liver cancer. In addition, Resolvins enhance tumor cell debris clearance by macrophages in the tumor's microenvironment. Resolvins, with their unique stereochemical structure, receptors, and biosynthetic pathways, provide a novel therapeutical approach to activating resolution mechanisms during cancer progression.

3.
Nanoscale Horiz ; 9(2): 295-304, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38086653

ABSTRACT

Cancer immunotherapy has received increasing attention in tumor therapy. However, insufficient infiltration of T cells and over-expressed PD-L1 checkpoint in tumor cells severely impede cancer immunotherapy. Here, an injectable hydrogel was designed to reinforce T cell infiltration and inactivate PD-L1 for powerful cancer immunotherapy. The hydrogel was created by sodium alginate (SA) as the gelator, where linagliptin particles and BMS-202 particles were present in hydrogel micropores. After gelation in the tumor site, the linagliptin powerfully suppressed chemokine CXCL10 degradation, enabling the introduced CXCL10 to realize sustainable chemotaxis towards strong T cell infiltration. Meanwhile, the BMS-202 inactivated PD-L1 of tumor cells, thereby eliminating the PD-L1-governed immune evasion. Therefore, the hydrogel in combination with CXCL10 demonstrated powerful cancer immunotherapy against primary and distant tumors, along with efficient inhibition of lung metastasis. Our study not only offers a potent platform against tumors, but also provides a conceptually new approach to reinforce cancer immunotherapy.


Subject(s)
Lung Neoplasms , T-Lymphocytes , Humans , T-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , Hydrogels , Immune Evasion , Linagliptin , Lung Neoplasms/therapy , Immunotherapy
4.
Acta Biomater ; 169: 306-316, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37574158

ABSTRACT

Prophylactic tumor vaccines hold great promise against tumor occurrence. However, their clinical efficacy remains low due to inadequate activation of strong-sustainable immunity. Herein, a biomembrane hydrogel was designed as a powerful single-shot prophylactic tumor vaccine. Mannose-decorated hybrid biomembrane (MHCM) modified with oxidized sodium alginate (OSA) was designed as a gelator (O-MHCM), where the hybrid biomembrane (HCM) is a hybridization of bacterial outer membrane vesicles (OMV) and tumor cell membranes (TCM). The O-MHCM enables quick gelation subcutaneously where the cysteine protease inhibitor E64 is encapsulated in hydrogel micropores. After a single vaccination of E64@O-MHCM hydrogel, MHCM and E64 are released sustainably due to OSA moiety degradation. The MHCM enables active targeting to dendritic cells (DC) and effective DC maturation. Meanwhile, the E64 enables sufficient antigen availability for subsequent cross presentation. Ultimately, strong and sustainable T lymphocyte-mediated immunity was elicited, demonstrating a strong prophylactic effect against breast tumors. This study provides a long-lasting platform to prevent tumor occurrence, opening an innovative avenue for the design of a single-shot prophylactic tumor vaccine. STATEMENT OF SIGNIFICANCE: Developing a single-shot prophylactic tumor vaccine to elicit strong-sustainable immunity is of great interest clinically. Here, a prophylactic tumor vaccine was designed using an injectable biomembrane hydrogel for achieving strong-sustainable immunity. The mannose-tailored hybrid biomembrane was modified with oxidized sodium alginate to result in a gelator, which enabled the formation of the hydrogel after subcutaneous injection. Cysteine protease inhibitor E64 was incorporated into the micropores of the hydrogel. The hydrogel induced strong-sustainable immunity through the continuous release of active components. This was facilitated by the mannose moiety, which enabled active targeting, as well as the antigen and adjuvant function of biomembrane, and the E64-enabled suppression of antigen degradation. The biomembrane hydrogel demonstrated powerful prevention of 4T1 breast tumors. This study offers an attractive strategy for designing a single-shot prophylactic tumor vaccine.


Subject(s)
Breast Neoplasms , Cancer Vaccines , Humans , Female , Hydrogels/pharmacology , Mannose , T-Lymphocytes , Antigens , Breast Neoplasms/drug therapy , Dendritic Cells
5.
J Control Release ; 358: 345-357, 2023 06.
Article in English | MEDLINE | ID: mdl-37150404

ABSTRACT

T cell-based immunotherapy (TCBI) is an emerging approach to combat tumors. However, the outcome of TCBI is still far from satisfaction clinically, owing to stumbling blocks from insufficient immunogenicity, T cell exhaustion and immune evasion from programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) pathway. Herein, an injectable tumor lysates-constructed hydrogel is reported to address these issues. Chemically modified tumor lysates are, for the first time, designed as the gelator to intratumorally construct hydrogel, achieving a robust antigen reservoir to induce strong immunogenicity. Meanwhile, hydrogel-encapsulated nicotinamide riboside and SB415286 enable strong mitophagy in T cells to prevent their exhaustion as well as powerfully genetical suppression of PD-1 expression to regulate immune evasion. Thus, our injectable hydrogel creates a robust immune niche within tumor, enabling to significantly potentiate TCBI. Our strategy pharmacologically regulates body's own T cells in situ, demonstrating potent immunotherapeutic effects and offering a conceptually new approach for TCBI.


Subject(s)
Hydrogels , Neoplasms , Humans , Programmed Cell Death 1 Receptor , T-Lymphocytes/metabolism , Immunotherapy , Tumor Microenvironment
6.
Acta Biomater ; 159: 300-311, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36642338

ABSTRACT

Ferroptosis has received increasing attentions in cancer therapy owing to its unique advantages over apoptosis. However, ferroptosis is governed by the efficiency of reactive oxygen species (ROS) production and the tumor cell antioxidant microenvironment that compromises therapeutic efficacy of ferroptosis. It is of great significance to develop a strategy that can both achieve high-efficiency ROS production and modulate tumor cell antioxidant microenvironment to amplify ferroptosis. However, until now, such a strategy has rarely been realized. Here, we, for the first time, reported a radiotherapy -mediated redox homeostasis-controllable nanomedicine for amplifying ferroptosis sensitivity in tumor therapy. The nanomedicine is constructed by co-assembling a ferroptosis inducer hemin and a thioredoxin 1 (Trx-1) inhibitor 1-methylpropyl 2-imidazolyl disulfide (PX-12) with human serum albumin. For our nanomedicine, hemin converts H2O2 to ROS via Fenton reaction to induce ferroptosis while PX-12 effectively inhibits the activity of antioxidant Trx-1 to suppress ROS depletion, resulting in amplified ferroptosis. Particularly, combining radiotherapy with the nanomedicine, radiotherapy depletes the other key antioxidant glutathione and generates additional radiotherapy-induced ROS, further boosting the ferroptosis effect. Therefore, our strategy can simultaneously ensure efficient ROS production and regulation of tumor cell antioxidant microenvironment, thereby enhancing efficacy of ferroptosis in tumor therapy. Our work offers an innovative approach to amplify ferroptosis sensitivity against tumors by simultaneously promoting ROS production and regulating redox homeostasis. STATEMENT OF SIGNIFICANCE: The antioxidants such as thioredoxin 1 (Trx-1) and glutathione (GSH) in tumor cells, are significantly upregulated by the innate cancer cellular redox homeostasis, severely restricting the reactive oxygen species (ROS)-based therapy and compromising the effect of Fenton reaction-induced ferroptosis against tumors. It is urgent to develop a strategy to simultaneously achieve Fenton reaction-induced ferroptosis and regulate the cancer cellular redox homeostasis against upregulated levels of Trx-1 and GSH. A radiotherapy-mediated redox homeostasis-regulatable nanomedicine was designed for amplifying ferroptosis sensitivity in tumor therapy, where the therapeutic efficacy of ferroptosis against tumors can be significantly amplified by integrating Fenton reaction-induced and radiotherapy-induced ferroptosis as well as PX-12-enabled inhibition of antioxidant Trx-1 and radiotherapy-induced downregulation of antioxidant GSH levels.


Subject(s)
Ferroptosis , Neoplasms , Humans , Antioxidants/pharmacology , Reactive Oxygen Species , Nanomedicine , Hemin/pharmacology , Hydrogen Peroxide/pharmacology , Oxidation-Reduction , Glutathione/metabolism , Homeostasis , Thioredoxins/metabolism , Thioredoxins/pharmacology , Cell Line, Tumor , Tumor Microenvironment
7.
Small ; 18(32): e2202663, 2022 08.
Article in English | MEDLINE | ID: mdl-35843879

ABSTRACT

T cell exhaustion caused by mitochondrial dysfunction is the major obstacle of T cells-based cancer immunotherapy. Besides exhausted T cells, the insufficient major histocompatibility complex class I (MHC I) on tumor cells leads to inefficient T cell recognition of tumor cells, compromising therapeutic efficacy. Therapeutic platform to regulate T cell exhaustion and MHC I expression for boosting T cells-based cancer immunotherapy has not been realized up to date. Herein, an injectable hydrogel is designed to simultaneously tune T cell exhaustion and MHC I expression for amplified cancer immunotherapy. The hydrogel is in situ constructed in tumor site by utilizing oxidized sodium alginate-modified tumor cell membrane vesicle (O-TMV) as a gelator, where axitinib is encapsulated in the lipid bilayer of O-TMV while 4-1BB antibody and proprotein convertase subtilisin/kexin type 9 inhibitor PF-06446846 nanoparticles are present in the cavities of hydrogel. After immune response trigged by O-TMV antigen, the 4-1BB antibody-promoted T cell mitochondrial biogenesis and the axitinib-lowered hypoxia synergistically reverse T cell exhaustion while the PF-06446846-amplified MHC I expression facilitates T cell recognition of tumor cells, demonstrating a powerful immunotherapeutic efficacy. This strategy on reprograming T cell exhaustion and improving T cell potency offers new concept for T cells-based cancer immunotherapy.


Subject(s)
Neoplasms , T-Lymphocytes , Antibodies , Axitinib , Histocompatibility Antigens Class I , Humans , Hydrogels , Immunotherapy , Neoplasms/therapy
8.
J Immunother Cancer ; 10(3)2022 03.
Article in English | MEDLINE | ID: mdl-35236742

ABSTRACT

BACKGROUND: On the basis of efficacy in mouse tumor models, multiple CD137 (4-1BB) agonist agents are being preclinically and clinically developed. The costimulatory molecule CD137 is inducibly expressed as a transmembrane or as a soluble protein (sCD137). Moreover, the CD137 cytoplasmic signaling domain is a key part in approved chimeric antigen receptors (CARs). Reliable pharmacodynamic biomarkers for CD137 ligation and costimulation of T cells will facilitate clinical development of CD137 agonists in the clinic. METHODS: We used human and mouse CD8 T cells undergoing activation to measure CD137 transcription and protein expression levels determining both the membrane-bound and soluble forms. In tumor-bearing mice plasma sCD137 concentrations were monitored on treatment with agonist anti-CD137 monoclonal antibodies (mAbs). Human CD137 knock-in mice were treated with clinical-grade agonist anti-human CD137 mAb (Urelumab). Sequential plasma samples were collected from the first patients intratumorally treated with Urelumab in the INTRUST clinical trial. Anti-mesothelin CD137-encompassing CAR-transduced T cells were stimulated with mesothelin coated microbeads. sCD137 was measured by sandwich ELISA and Luminex. Flow cytometry was used to monitor CD137 surface expression. RESULTS: CD137 costimulation upregulates transcription and protein expression of CD137 itself including sCD137 in human and mouse CD8 T cells. Immunotherapy with anti-CD137 agonist mAb resulted in increased plasma sCD137 in mice bearing syngeneic tumors. sCD137 induction is also observed in human CD137 knock-in mice treated with Urelumab and in mice transiently humanized with T cells undergoing CD137 costimulation inside subcutaneously implanted Matrigel plugs. The CD137 signaling domain-containing CAR T cells readily released sCD137 and acquired CD137 surface expression on antigen recognition. Patients treated intratumorally with low dose Urelumab showed increased plasma concentrations of sCD137. CONCLUSION: sCD137 in plasma and CD137 surface expression can be used as quantitative parameters dynamically reflecting therapeutic costimulatory activity elicited by agonist CD137-targeted agents.


Subject(s)
Immunotherapy , Neoplasms , Animals , Biomarkers/metabolism , CD8-Positive T-Lymphocytes , Humans , Mice , Neoplasms/drug therapy , Receptors, Tumor Necrosis Factor
9.
Front Oncol ; 12: 809068, 2022.
Article in English | MEDLINE | ID: mdl-35311098

ABSTRACT

Primary splenic angiosarcoma (PSA) is a rare malignancy with poor prognosis. At present, little study is available on immunotherapy in PSA. Here, we report a case of a patient with metastatic PSA who was treated with programmed death-1 (PD-1) inhibitors and vascular endothelial growth factor (VEGF) tyrosine kinase inhibitors combined therapy and achieved complete response (CR). The patient was a 57-year-old woman with three liver metastases. She was treated with seven cycles of toripalimab plus anlotinib. Programmed death-ligand 1 (PD-L1) immunohistochemistry and next-generation sequencing was performed, and the PD-L1 tumor proportion score was 75%. Finally, she achieved CR after six cycles of the combined therapy regimen. No serious adverse events were detected. To the best of our knowledge, this is the first clinical evidence that anti-PD-1 plus anti-VEGF therapy might be a promising option for patients with metastatic PSA. However, more clinical trials are needed to verify this conclusion.

10.
Cancer Discov ; 11(7): 1700-1715, 2021 07.
Article in English | MEDLINE | ID: mdl-33658301

ABSTRACT

Specific mechanisms by which tumor-infiltrating lymphocytes (TIL) become dysfunctional remain poorly understood. Here, we employed a two-pronged approach using single-cell mass cytometry and tissue imaging technologies to dissect TILs from 25 patients with resectable and 35 patients with advanced non-small cell lung cancer (NSCLC). We identified a burned-out CD8+ TIL subset (Ebo) that specifically accumulated within the tumor microenvironment (TME) but not in adjacent nontumoral tissues. Ebo showed the highest expression of proliferation and activation markers but produced the lowest amount of IFNγ and were the most apoptotic CD8+ TIL subset. Using a humanized patient-derived tumor xenograft model, we demonstrated that Ebo expansion occurred within the TME in a PD-1/B7-H1 pathway-dependent manner. Ebo abundance in baseline tumor tissues was associated with resistance to anti-PD therapy in patients with NSCLC. Our study identifies a dysfunctional TIL subset, with distinct features from previously described exhausted T cells, and implies strategies to overcome immunotherapy resistance. SIGNIFICANCE: We identified a highly proliferative, overactivated, and apoptotic dysfunctional CD8+ tumor-infiltrating subpopulation that is functionally distinct from previously described exhausted T cells. This population is expanded in lung cancer tissues in a PD-1/B7-H1-dependent manner, and its abundance is associated with resistance to cancer immunotherapy, thus becoming a potential tissue biomarker.This article is highlighted in the In This Issue feature, p. 1601.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Tumor Microenvironment , Aged , Animals , Carcinoma, Non-Small-Cell Lung/therapy , Female , Humans , Immunotherapy , Lung Neoplasms/therapy , Male , Mice , Mice, Inbred NOD , Prospective Studies
11.
Sci Transl Med ; 11(522)2019 12 11.
Article in English | MEDLINE | ID: mdl-31826980

ABSTRACT

Systemic lupus erythematosus (SLE) and discoid lupus erythematosus (DLE) of the skin are autoimmune diseases characterized by inappropriate immune responses against self-proteins; the key elements that determine disease pathogenesis and progression are largely unknown. Here, we show that mice lacking immune inhibitory receptor VISTA or programmed death-1 homolog (PD-1H KO) on a BALB/c background spontaneously develop cutaneous and systemic autoimmune diseases resembling human lupus. Cutaneous lupus lesions of PD-1H KO mice have clustering of plasmacytoid dendritic cells (pDCs) similar to human DLE. Using mass cytometry, we identified proinflammatory neutrophils as critical early immune infiltrating cells within cutaneous lupus lesions of PD-1H KO mice. We also found that PD-1H is highly expressed on immune cells in human SLE, DLE lesions, and cutaneous lesions of MRL/lpr mice. A PD-1H agonistic monoclonal antibody in MRL/lpr mice reduces cutaneous disease, autoantibodies, inflammatory cytokines, chemokines, and immune cell expansion. Furthermore, PD-1H on both T cells and myeloid cells including neutrophils and pDCs could transmit inhibitory signals, resulting in reduced activation and function, establishing PD-1H as an inhibitory receptor on T cells and myeloid cells. On the basis of these findings, we propose that PD-1H is a critical element in the pathogenesis and progression of lupus, and PD-1H activation could be effective for treatment of systemic and cutaneous lupus.


Subject(s)
Autoimmunity , Lupus Erythematosus, Cutaneous/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Proteins/metabolism , Animals , Arthritis/pathology , Autoantibodies/immunology , Dendritic Cells/immunology , Humans , Inflammation/pathology , Interferon Type I/metabolism , Membrane Proteins/agonists , Membrane Proteins/deficiency , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Myeloid Cells/metabolism , Neutrophils/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Terpenes , Up-Regulation
12.
Nat Med ; 25(4): 656-666, 2019 04.
Article in English | MEDLINE | ID: mdl-30833750

ABSTRACT

Overexpression of the B7-H1 (PD-L1) molecule in the tumor microenvironment (TME) is a major immune evasion mechanism in some patients with cancer, and antibody blockade of the B7-H1/PD-1 interaction can normalize compromised immunity without excessive side-effects. Using a genome-scale T cell activity array, we identified Siglec-15 as a critical immune suppressor. While only expressed on some myeloid cells normally, Siglec-15 is broadly upregulated on human cancer cells and tumor-infiltrating myeloid cells, and its expression is mutually exclusive to B7-H1, partially due to its induction by macrophage colony-stimulating factor and downregulation by IFN-γ. We demonstrate that Siglec-15 suppresses antigen-specific T cell responses in vitro and in vivo. Genetic ablation or antibody blockade of Siglec-15 amplifies anti-tumor immunity in the TME and inhibits tumor growth in some mouse models. Taken together, our results support Siglec-15 as a potential target for normalization cancer immunotherapy.


Subject(s)
Immunoglobulins/metabolism , Immunotherapy , Membrane Proteins/metabolism , Neoplasms/immunology , Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Epitopes , Humans , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/metabolism , Neoplasms/pathology , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/immunology
13.
Cell Mol Immunol ; 15(10): 888-897, 2018 10.
Article in English | MEDLINE | ID: mdl-28479601

ABSTRACT

The role of B7-DC in T-cell responses remains controversial because both coinhibitory and costimulatory functions have been reported in various experimental systems in vitro and in vivo. In addition to interacting with the coinhibitory receptor PD-1, B7-DC has also been shown to bind repulsive guidance molecule b (RGMb). The functional consequences of the B7-DC/RGMb interaction, however, remain unclear. More than a decade ago, we reported that replacement of a murine B7-DC mutant lysine with serine (K113S) at positive 113 resulted in a loss of binding capacity to PD-1. Nevertheless, K113S remained costimulatory for T cells in vitro, implicating a dual functionality for B7-DC in T-cell responses. Here we show that recombinant K113S protein interacts with RGMb with a similar affinity to wild-type B7-DC. More importantly, K113S costimulates CD4+ T-cell responses via RGMb and promotes Th1 polarization. RGMb is expressed on the surface of naive mouse T cells, macrophages, neutrophils and dendritic cells. Finally, K113S/RGMb costimulation suppresses Th2-mediated asthma and ameliorates small airway inflammation and lung pathology in an experimental mouse model. Our findings indicate that RGMb is a costimulatory receptor for B7-DC. These findings from the K113S variant provide not only a possible explanation for the B7-DC-triggered contradictory effects on T-cell responses, but also a novel approach to investigate the B7-DC/PD-1/RGMb axis. Recombinant K113S or its derivatives could potentially be developed as an agonist for RGMb to costimulate the Th1 response without triggering PD-1-mediated T-cell inhibition.


Subject(s)
Asthma/immunology , Nerve Tissue Proteins/immunology , Programmed Cell Death 1 Ligand 2 Protein/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Amino Acid Substitution , Animals , Asthma/genetics , Asthma/pathology , CHO Cells , Cell Adhesion Molecules, Neuronal , Cricetulus , Female , GPI-Linked Proteins , Humans , Mice , Mice, Inbred BALB C , Mutation, Missense , Nerve Tissue Proteins/genetics , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Th1 Cells/pathology , Th2 Cells/pathology
14.
15.
PLoS One ; 11(12): e0168384, 2016.
Article in English | MEDLINE | ID: mdl-28030551

ABSTRACT

Mulching management has been used in many places all over the world to improve agricultural sustainability. However, the cycling of carbon in the soil under applications of mulch on sloping arable land is not yet fully understood. A four-year field experiment was carried out in Xiaofuling watershed of Danjiangkou reservoir in China. The object was to evaluate the effects of the application of straw mulch (ST) and grass mulch (GT) on dynamic changes in soil organic carbon and its fractions. Results showed that mulch applied on the soil surface increased the contents of SOC and its active fractions in the soil. Compared to the control without cover (CK), ST and GT treatments increased the contents of SOC, LOC, DOC, POC and EOC by 14.73%, 16.5%, 22.5%, 41.5% and 21%, respectively, in the 0-40 cm soil layer, and by 17%, 14%, 19%, and 30%, respectively, in the 0-100 cm soil layer. The contents of organic carbon and its active fractions decreased with increasing soil depth in all of the treatments. SOC was accumulated in the period of December to the following March. The contents of soil DOC and LOC were high in January to March, while the contents of soil POC and EOC were high in June to September. The relative contents of soil organic carbon fractions were POC > EOC > LOC > DOC over the four years. Straw mulching had no significant effect on the changes in soil organic carbon active fractions during the different periods. Based on this long-term field experiment in Danjiangkou reservoir, we found that straw mulching had a significant effect on soil, increasing SOC content and stock in slopping arable land, and that live grass mulching was more effective than rice straw mulching. We discuss possible optimal periods for the implementation of mulching practices on sloping land.


Subject(s)
Agriculture/methods , Carbon Sequestration , Carbon/chemistry , Citrus/growth & development , Soil/chemistry , Water Movements
16.
Toxicol Sci ; 145(1): 118-27, 2015 May.
Article in English | MEDLINE | ID: mdl-25666835

ABSTRACT

Endosulfan as a new member of persistent organic pollutants has been shown to induce reproductive dysfunction in various animal models. However, the action mechanism of endosulfan-produced reproductive toxicity remains largely unknown. This study was focused on investigating the reproductive toxicity induced by α-endosulfan and clarifying the role of mitochondria and genotoxic response genes in germ cell apoptosis of Caenorhabditis elegans. Our data showed that endosulfan induced a dose-dependent decrease of life span, fecundity, and hatchability, whereas the germ cell apoptosis was dose-dependently increased. The mitochondria membrane potential was disrupted by endosulfan, leading to a significant increase of germ cell apoptosis in mev-1(kn-1) mutant. However, the apoptotic effects of endosulfan were blocked in mutants of cep-1(w40), egl-1(n487), and hus-1(op241), indicating conserved genotoxic response genes played an essential role in endosulfan-induced germ cell apoptosis. Furthermore, exposure to endosulfan induced the accumulation of HUS-1::GFP foci and the germ cell cycle arrest. These findings provided clear evidence that endosulfan caused significant adverse effects on the reproduction system of C. elegans and increased germ cell apoptosis, which was regulated by mitochondrial dysfunction and DNA damage response genes. This study may help to understand the signal transduction pathways involved in endosulfan-induced reproductive toxicity.


Subject(s)
Apoptosis/drug effects , Caenorhabditis elegans/drug effects , Endosulfan/toxicity , Germ Cells/drug effects , Mutagens/toxicity , Reproduction/drug effects , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Germ Cells/cytology
17.
J Infect Dis ; 210(1): 89-98, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24403554

ABSTRACT

miR-155 (microRNA-155) is an important noncoding RNA in regulating host inflammatory responses. However, its regulatory role in ocular infection remains unclear. Our study first explored the function of miR-155 in Pseudomonas aeruginosa-induced keratitis, one of the most common sight-threatening ocular diseases. We found that miR-155 expression was enhanced in human and mouse corneas after P. aeruginosa infection and was mainly expressed in macrophages but not neutrophils. In vivo studies demonstrated that miR-155 knockout mice displayed more resistance to P. aeruginosa keratitis, with a higher inducible nitric oxide synthase level and a lower bacterial burden. More importantly, in vitro data indicated that miR-155 suppressed the macrophage-mediated bacterial phagocytosis and intracellular killing of P. aeruginosa by targeting Rheb (Ras homolog enriched in brain). To the best of our knowledge, this is the first study to explore the role of miR-155 in bacterial keratitis, which may provide a promising target for clinical treatment of P. aeruginosa keratitis and other infectious diseases.


Subject(s)
Keratitis/immunology , Keratitis/microbiology , MicroRNAs/metabolism , Monomeric GTP-Binding Proteins/immunology , Neuropeptides/immunology , Pseudomonas aeruginosa/immunology , Adult , Animals , Bacterial Load , Female , Humans , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , Middle Aged , Nitric Oxide/metabolism , Phagocytosis , Ras Homolog Enriched in Brain Protein , Young Adult
18.
J Infect ; 67(6): 584-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911965

ABSTRACT

OBJECTIVE: To explore the role of ß-catenin in Pseudomonas aeruginosa (PA) keratitis. METHODS: Western-blot and immunostaining assay were used to determine the ß-catenin protein expression in C57BL/6 (B6) corneas and in in vitro cultured murine cells including macrophage-like RAW264.7 cells, bone marrow-derived neutrophils and A6(1) corneal epithelial cells. B6 mice were subconjunctivally injected with lentivirus expressing active mutant of ß-catenin (ß-cat-lentivirus) vs appropriate control (Ctl-lentivirus), and then infected with PA. Pro-inflammatory cytokine levels were examined using real-time PCR and ELISA, and bacterial burden was assessed using plate count assays both in vivo and in vitro. RESULTS: ß-Catenin protein expression was decreased in B6 corneas, murine macrophage-like RAW264.7 cells, mouse bone marrow-derived neutrophils and mouse A6(1) corneal epithelial cells after PA infection. Over-expression of ß-catenin in B6 corneas significantly reduced the severity of corneal disease after PA infection, by decreasing pro-inflammatory cytokine expression and bacterial burden. In vitro data further demonstrated that over-expression of ß-catenin suppressed pro-inflammatory cytokine production but enhanced bacterial clearance in macrophages and neutrophils. CONCLUSIONS: ß-Catenin reduces the severity of PA keratitis by decreasing corneal inflammation and bacterial burden.


Subject(s)
Keratitis/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , beta Catenin/immunology , Animals , Cell Line , Cornea/cytology , Cornea/immunology , Disease Progression , Epithelial Cells/immunology , Female , Keratitis/microbiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Pseudomonas Infections/microbiology
19.
Mol Vis ; 19: 1502-14, 2013.
Article in English | MEDLINE | ID: mdl-23878501

ABSTRACT

PURPOSE: To explore the role of lithium chloride (LiCl) in Pseudomonas aeruginosa (PA) keratitis. METHODS: B6 mice were subconjunctivally injected with LiCl in contrast to appropriate control sodium chloride (NaCl), and then routinely infected with PA. Clinical score, slit-lamp photography, hematoxylin and eosin (H&E) staining, and bacterial plate counts were used to determine the role of LiCl in PA keratitis. Messenger ribonucleic acid and protein levels of inflammatory cytokines in PA-challenged mouse corneas and in vitro cultured macrophages and neutrophils were measured with real-time PCR and enzyme-linked immunosorbent assay (ELISA), respectively. Apoptosis of the infiltrating inflammatory cells in the PA-infected murine corneas was assessed using terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling staining and propidium iodide staining associated with flow cytometry. In cultured murine macrophages and neutrophils, cell apoptosis was determined with annexin V/propidium iodide double staining associated with flow cytometry and western blot analysis for cleaved caspase-3 and cleaved poly(ADP-ribose) polymerase. RESULTS: Treatment with LiCl reduced the severity of corneal disease by reducing corneal inflammatory response and bacterial burden. Moreover, LiCl increased anti-inflammatory cytokine interleukin-10 levels, decreased proinflammatory cytokine tumor necrosis factor-α levels, and enhanced apoptosis of infiltrating macrophages and neutrophils in the PA-infected mouse corneas. In vitro studies further confirmed that LiCl elevated anti-inflammatory cytokine expression but reduced proinflammatory cytokine production, as well as promoted cell apoptosis in murine macrophages and neutrophils. CONCLUSIONS: This study demonstrates a protective role of LiCl in PA keratitis. LiCl promotes host resistance against PA infection by suppressing inflammatory responses, enhancing inflammatory cell apoptosis, and promoting bacterial clearance.


Subject(s)
Disease Resistance/drug effects , Host-Pathogen Interactions/drug effects , Keratitis/immunology , Keratitis/microbiology , Lithium Chloride/pharmacology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/drug effects , Animals , Apoptosis/drug effects , Cornea/drug effects , Cornea/microbiology , Cornea/pathology , Cytokines/metabolism , Disease Resistance/immunology , Female , Flow Cytometry , Host-Pathogen Interactions/immunology , Inflammation Mediators/metabolism , Keratitis/drug therapy , Lithium Chloride/therapeutic use , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Protective Agents/therapeutic use , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology
20.
Invest Ophthalmol Vis Sci ; 54(5): 3451-62, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23611998

ABSTRACT

PURPOSE: To explore the role of triggering receptor expressed on myeloid cells 2 (TREM-2) in Pseudomonas aeruginosa (PA) keratitis. METHODS: BALB/c mice were routinely infected with PA and evaluated at various postinfection time points for corneal expression of TREM-2, by real-time PCR, Western blot, and flow cytometry. Next, BALB/c and C57BL/6 mice were respectively treated with TREM-2 siRNA or agonistic anti-TREM-2 antibody, to determine the role of TREM-2 in PA keratitis. Bacterial load and neutrophil infiltration were tested by plate count and myeloperoxidase assay, respectively. Th1-/Th2-type and proinflammatory cytokine expression were tested by real-time PCR and ELISA after in vivo and in vitro silencing of TREM-2. Moreover, phosphorylated Akt levels were tested by Western blot in murine macrophages after treatment with agonistic anti-TREM-2 antibody. mRNA levels of proinflammatory cytokines were examined in murine macrophages after TREM-2 activation and lipopolysaccharide stimulation, following pretreatment with inhibitors for PI3K or Akt, to determine whether PI3K/Akt is required in TREM-2-mediated immune modulation. In addition, BALB/c mice were treated with wortmannin and analyzed for bacterial load and proinflammatory cytokine expression. RESULTS: TREM-2 expression was elevated in the infected BALB/c corneas at 3 or 5 days postinfection. Silencing of TREM-2 accelerated disease progression by enhancing bacterial load and corneal inflammation, whereas activation of TREM-2 promoted host resistance to PA keratitis. PI3K/Akt signaling is required in the TREM-2-mediated immune modulation, and inhibition of PI3K resulted in worsened disease after PA corneal infection. CONCLUSIONS: TREM-2 promoted host resistance to PA infection by suppressing corneal inflammation via activation of the PI3K/Akt pathway.


Subject(s)
Eye Infections, Bacterial/prevention & control , Immunity, Innate , Keratitis/prevention & control , Membrane Glycoproteins/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pseudomonas Infections/prevention & control , Receptors, Immunologic/physiology , Animals , Blotting, Western , Cells, Cultured , Colony Count, Microbial , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Eye Infections, Bacterial/immunology , Eye Infections, Bacterial/microbiology , Flow Cytometry , Keratitis/immunology , Keratitis/microbiology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/physiology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...