Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37745595

ABSTRACT

The tumor microenvironment (TME) is characterized by a network of cancer cells, recruited immune cells and extracellular matrix (ECM) in a hypoxic microenvironment. However, the specific role of neutrophils during tumor development, and their interactions with other immune cells is still not well understood. Thus, there is a need to investigate the interaction between primary neutrophils and natural killer cells and the resulting effects on tumor development. Here we use both standard well plate culture and an under oil microfluidic (UOM) assay with an integrated extracellular cell matrix (ECM) bridge to elucidate how naive primary neutrophils respond to both patient derived tumor cells and tumor cell lines. Our data demonstrated that both patient derived head and neck squamous cell carcinoma (HNSCC) tumor cells and MDA-MB-231 breast cancer cells trigger cluster formation in neutrophils, and the swarm of neutrophils restricts tumor invasion through the generation of reactive oxygen species (ROS) and neutrophil extracellular trap (NETs) release within the neutrophil cluster. However, we also observed that the presence of neutrophils downregulates granzyme B in NK-92 cells and the resulting NETs can obstruct NK cells from penetrating the tumor mass in vitro suggesting a dual role for neutrophils in the TME. Further, using label-free optical metabolic imaging (OMI) we observed changes in the metabolic activities of primary neutrophils during the different swarming phases when challenged with tumor cells. Finally, our data demonstrates that neutrophils in direct contact, or in close proximity, with tumor cells exhibit greater metabolic activities (lower nicotinamide adenine dinucleotide phosphate (NAD(P)H) mean lifetime) compared to non-contact neutrophils.

3.
bioRxiv ; 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37873450

ABSTRACT

Acute respiratory distress syndrome due to non-pulmonary causes exhibits prominent endothelial activation which is challenging to assess in critically ill patients. Preclinical in vivo models of sepsis and ARDS have failed to yield useful therapies in humans, perhaps due to interspecies differences in inflammatory responses. Use of microphysiological systems (MPS) offer improved fidelity to human biological responses and better predict pharmacological responses than traditional culture. We adapted a lung endothelial MPS based on the LumeNEXT platform to evaluate the effect of plasma from critically ill sepsis patients on endothelial permeability, adhesion molecule expression and inflammatory cytokine production. Lumens incubated with sepsis plasma exhibited areas of contraction, loss of cellular coverage, and luminal defects. Sepsis plasma-incubated lumens had significantly increased permeability compared to lumens incubated with healthy donor plasma. ICAM-1 expression increased significantly in lumens incubated with sepsis plasma compared with those incubated with healthy control plasma, while concentrations of IL-6, IL-18, and soluble VEGF-R1 increased in sepsis plasma before and after incubation in the MPS compared with healthy control plasma. Use of the lung endothelial MPS may enable interrogation of specific mechanisms of endothelial dysfunction that promote ARDS in sepsis patients.

4.
Nat Commun ; 14(1): 6681, 2023 10 21.
Article in English | MEDLINE | ID: mdl-37865647

ABSTRACT

Numerous studies are exploring the use of cell adoptive therapies to treat hematological malignancies as well as solid tumors. However, there are numerous factors that dampen the immune response, including viruses like human immunodeficiency virus. In this study, we leverage human-derived microphysiological models to reverse-engineer the HIV-immune system interaction and evaluate the potential of memory-like natural killer cells for HIV+ head and neck cancer, one of the most common tumors in patients living with human immunodeficiency virus. Here, we evaluate multiple aspects of the memory-like natural killer cell response in human-derived bioengineered environments, including immune cell extravasation, tumor penetration, tumor killing, T cell dependence, virus suppression, and compatibility with retroviral medication. Overall, these results suggest that memory-like natural killer cells are capable of operating without T cell assistance and could simultaneously destroy head and neck cancer cells as well as reduce viral latency.


Subject(s)
HIV Infections , Head and Neck Neoplasms , Viruses , Humans , HIV , Killer Cells, Natural , Immunotherapy/methods
5.
Biomaterials ; 298: 122136, 2023 07.
Article in English | MEDLINE | ID: mdl-37178589

ABSTRACT

Regional metastasis of head and neck cancer (HNC) is prevalent (approximately 50% of patients at diagnosis), yet the underlying drivers and mechanisms of lymphatic spread remain unclear. The complex tumor microenvironment (TME) of HNC plays a crucial role in disease maintenance and progression; however, the contribution of the lymphatics remains underexplored. We created a primary patient cell derived microphysiological system that incorporates cancer-associated-fibroblasts from patients with HNC alongside a HNC tumor spheroid and a lymphatic microvessel to create an in vitro TME platform to investigate metastasis. Screening of soluble factor signaling identified novel secretion of macrophage migration inhibitory factor (MIF) by lymphatic endothelial cells conditioned in the TME. Importantly, we also observed patient-to-patient heterogeneity in cancer cell migration similar to the heterogeneity observed in clinical disease. Optical metabolic imaging at the single cell level identified a distinct metabolic profile of migratory versus non-migratory HNC cells in a microenvironment dependent manner. Additionally, we report a unique role of MIF in increasing HNC reliance on glycolysis over oxidative phosphorylation. This multicellular, microfluidic platform expands the tools available to explore HNC biology in vitro through multiple orthogonal outputs and establishes a system with enough resolution to visualize and quantify patient-to-patient heterogeneity.


Subject(s)
Head and Neck Neoplasms , Macrophage Migration-Inhibitory Factors , Humans , Macrophage Migration-Inhibitory Factors/metabolism , Monocytes/metabolism , Endothelial Cells/metabolism , Cell Movement , Cell Line, Tumor , Tumor Microenvironment
6.
Integr Biol (Camb) ; 152023 04 11.
Article in English | MEDLINE | ID: mdl-36781971

ABSTRACT

The lymphatic system plays an active role during infection, however the role of lymphatic-neutrophil interactions in host-defense responses is not well understood. During infection with pathogens such as Pseudomonas aeruginosa, Staphylococcus aureus and Yersinia pestis, neutrophils traffic from sites of infection through the lymphatic vasculature, to draining lymph nodes to interact with resident lymphocytes. This process is poorly understood, in part, due to the lack of in vitro models of the lymphatic system. Here we use a 3D microscale lymphatic vessel model to examine neutrophil-lymphatic cell interactions during host defense responses to pathogens. In previous work, we have shown that follistatin is secreted at high concentrations by lymphatic endothelial cells during inflammation. Follistatin inhibits activin A, a member of the TGF-ß superfamily, and, together, these molecules form a signaling pathway that plays a role in regulating both innate and adaptive immune responses. Although follistatin and activin A are constitutively produced in the pituitary, gonads and skin, their major source in the serum and their effects on neutrophils are poorly understood. Here we report a microfluidic model that includes both blood and lymphatic endothelial vessels, and neutrophils to investigate neutrophil-lymphatic trafficking during infection with P. aeruginosa. We found that lymphatic endothelial cells produce secreted factors that increase neutrophil migration toward P. aeruginosa, and are a significant source of both follistatin and activin A during Pseudomonas infection. We determined that follistatin produced by lymphatic endothelial cells inhibits activin A, resulting in increased neutrophil migration. These data suggest that the follistatin:activin A ratio influences neutrophil trafficking during infection with higher ratios increasing neutrophil migration.


Subject(s)
Follistatin , Pseudomonas aeruginosa , Follistatin/metabolism , Pseudomonas aeruginosa/metabolism , Neutrophils/metabolism , Endothelium, Lymphatic/metabolism , Endothelial Cells/metabolism
7.
Sci Adv ; 8(18): eabm8012, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35544643

ABSTRACT

Protozoan parasites that infect humans are widespread and lead to varied clinical manifestations, including life-threatening illnesses in immunocompromised individuals. Animal models have provided insight into innate immunity against parasitic infections; however, species-specific differences and complexity of innate immune responses make translation to humans challenging. Thus, there is a need for in vitro systems that can elucidate mechanisms of immune control and parasite dissemination. We have developed a human microphysiological system of intestinal tissue to evaluate parasite-immune-specific interactions during infection, which integrates primary intestinal epithelial cells and immune cells to investigate the role of innate immune cells during epithelial infection by the protozoan parasite, Toxoplasma gondii, which affects billions of people worldwide. Our data indicate that epithelial infection by parasites stimulates a broad range of effector functions in neutrophils and natural killer cell-mediated cytokine production that play immunomodulatory roles, demonstrating the potential of our system for advancing the study of human-parasite interactions.


Subject(s)
Parasites , Toxoplasma , Animals , Host-Parasite Interactions , Humans , Immunity, Innate , Neutrophils
8.
Cancers (Basel) ; 14(3)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35159026

ABSTRACT

Bone metastases represent a lethal condition that frequently occurs in solid tumors such as prostate, breast, lung, and renal cell carcinomas, and increase the risk of skeletal-related events (SREs) including pain, pathologic fractures, and spinal cord compression. This unique metastatic niche consists of a multicellular complex that cancer cells co-opt to engender bone remodeling, immune suppression, and stromal-mediated therapeutic resistance. This review comprehensively discusses clinical challenges of bone metastases, novel preclinical models of the bone and bone marrow microenviroment, and crucial signaling pathways active in bone homeostasis and metastatic niche. These studies establish the context to summarize the current state of investigational agents targeting BM, and approaches to improve BM-targeting therapies. Finally, we discuss opportunities to advance research in bone and bone marrow microenvironments by increasing complexity of humanized preclinical models and fostering interdisciplinary collaborations to translational research in this challenging metastatic niche.

9.
Front Immunol ; 12: 781535, 2021.
Article in English | MEDLINE | ID: mdl-34899746

ABSTRACT

During infection, neutrophils are the most abundantly recruited innate immune cells at sites of infection, playing critical roles in the elimination of local infection and healing of the injury. Neutrophils are considered to be short-lived effector cells that undergo cell death at infection sites and in damaged tissues. However, recent in vitro and in vivo evidence suggests that neutrophil behavior is more complex and that they can migrate away from the inflammatory site back into the vasculature following the resolution of inflammation. Microfluidic devices have contributed to an improved understanding of the interaction and behavior of neutrophils ex vivo in 2D and 3D microenvironments. The role of reverse migration and its contribution to the resolution of inflammation remains unclear. In this review, we will provide a summary of the current applications of microfluidic devices to investigate neutrophil behavior and interactions with other immune cells with a focus on forward and reverse migration in neutrophils.


Subject(s)
Cell Communication , Chemotaxis, Leukocyte , Inflammation Mediators/metabolism , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Neutrophils/metabolism , Animals , Cells, Cultured , Host-Pathogen Interactions , Humans , Neutrophils/immunology , Neutrophils/pathology , Phenotype , Signal Transduction
10.
Integr Biol (Camb) ; 13(11): 259-268, 2021 12 30.
Article in English | MEDLINE | ID: mdl-34931665

ABSTRACT

Renal cell carcinoma (RCC) is the third most common genitourinary cancer in the USA. Despite recent advances in the treatment for advanced and metastatic clear cell RCC (ccRCC), the 5-year relative survival rate for the distant disease remains at 12%. Cabozantinib, a tyrosine kinase inhibitor (TKI), which is one of the first-line therapies approved to treat advanced ccRCC as a single agent, is now being investigated as a combination therapy with newer immunotherapeutic agents. However, not much is known about how cabozantinib modulates the immune system. Here, we present a high throughput tri-culture model that incorporates cancer cells, endothelial cells, and patient-derived immune cells to study the effect of immune cells from patients with ccRCC on angiogenesis and cabozantinib resistance. We show that circulating immune cells from patients with ccRCC induce cabozantinib resistance via increased secretion of a set of pro-angiogenic factors. Using multivariate partial least square regression modeling, we identified CD4+ T cell subsets that are correlated with cabozantinib resistance and report the changes in the frequency of these populations in ccRCC patients who are undergoing cabozantinib therapy. These findings provide a potential set of biomarkers that should be further investigated in the current TKI-immunotherapy combination clinical trials to improve personalized treatments for patients with ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Anilides/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/secondary , Endothelial Cells , Female , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Male , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines
11.
EBioMedicine ; 73: 103634, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34673450

ABSTRACT

BACKGROUND: In head and neck cancer, intratumour lymphatic density and tumour lymphangiogenesis have been correlated with lymphatic metastasis, making lymphangiogenesis a promising therapeutic target. However, inter-patient tumour heterogeneity makes it challenging to predict tumour progression and lymph node metastasis. Understanding the lymphangiogenic-promoting factors leading to metastasis (e.g., tumour-derived fibroblasts or TDF), would help develop strategies to improve patient outcomes. METHODS: A microfluidic in vitro model of a tubular lymphatic vessel was co-cultured with primary TDF from head and neck cancer patients to evaluate the effect of TDF on lymphangiogenesis. We assessed the length and number of lymphangiogenic sprouts and vessel permeability via microscopy and image analysis. Finally, we characterised lymphatic vessel conditioning by TDF via RT-qPCR. FINDINGS: Lymphatic vessels were conditioned by the TDF in a patient-specific manner. Specifically, the presence of TDF induced sprouting, altered vessel permeability, and increased the expression of pro-lymphangiogenic genes. Gene expression and functional responses in the fibroblast-conditioned lymphatic vessels were consistent with the patient tumour stage and lymph node status. IGF-1, upregulated among patients, was targeted to validate our personalised medicine approach. Interestingly, IGF-1 blockade was not effective across different patients. INTERPRETATION: The use of lymphatic organotypic models incorporating head and neck TDF provides insight into the pathways leading to lymphangiogenesis in each patient. This model provided a platform to test anti-angiogenic therapeutics and inform of their effectiveness for individual patients. FUNDING: NIH R33CA225281. Wisconsin Head and Neck SPORE NIH P50DE026787. NIH R01AI34749.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Head and Neck Neoplasms/pathology , Lymphangiogenesis , Neovascularization, Pathologic , Biomarkers , Cancer-Associated Fibroblasts/pathology , Cell Line , Coculture Techniques , Fluorescent Antibody Technique , Head and Neck Neoplasms/metabolism , Humans , Immunohistochemistry , Neovascularization, Pathologic/metabolism , Organoids
12.
Integr Biol (Camb) ; 12(10): 250-262, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33034643

ABSTRACT

The prostate tumor microenvironment (TME) is strongly immunosuppressive; it is largely driven by alteration in cell phenotypes (i.e. tumor-associated macrophages and exhausted cytotoxic T cells) that result in pro-tumorigenic conditions and tumor growth. A greater understanding into how these altered immune cell phenotypes are developed and could potentially be reversed would provide important insights into improved treatment efficacy for prostate cancer. Here, we report a microfluidic model of the prostate TME that mimics prostate ducts across various stages of prostate cancer progression, with associated stroma and immune cells. Using this platform, we exposed immune cells to a benign prostate TME or a metastatic prostate TME and investigated their metabolism, gene and cytokine expression. Immune cells exposed to the metastatic TME showed metabolic differences with a higher redox ratio indicating a switch to a more glycolytic metabolic profile. These cells also increased expression of pro-tumor response cytokines that have been shown to increase cell migration and angiogenesis such as Interleukin-1 (IL-1) a and Granulocyte-macrophage colony-stimulating factor (GM-CSF). Lastly, we observed decreased TLR, STAT signaling and TRAIL expression, suggesting that phenotypes derived from exposure to the metastatic TME could have an impaired anti-tumor response. This platform could provide a valuable tool for studying immune cell phenotypes in in vitro tumor microenvironments.


Subject(s)
Immune System , Prostatic Neoplasms/immunology , Prostatic Neoplasms/physiopathology , Tumor Microenvironment , Cell Movement , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Fibroblasts/metabolism , Glycolysis , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunosuppression Therapy , In Vitro Techniques , Leukocytes, Mononuclear/metabolism , Male , Microfluidics , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Organ Culture Techniques , Oxidation-Reduction , Phenotype , Prostate/metabolism , STAT Transcription Factors/metabolism , Signal Transduction
13.
Clin Exp Allergy ; 50(8): 904-914, 2020 08.
Article in English | MEDLINE | ID: mdl-32542913

ABSTRACT

BACKGROUND: Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is expressed on mast cells and eosinophils, but information about Siglec-8 expression and function in the lung is limited. A humanized antibody, AK002, targeting Siglec-8 is undergoing development for treatment of diseases associated with mast cell and eosinophil-driven inflammation. OBJECTIVE: To characterize Siglec-8 expression in the airway in asthma and determine whether antibodies that target Siglec-8 (S8mAbs) can decrease airway eosinophils in asthma or inhibit lung mast cell activation. METHODS: Gene expression profiling and flow cytometry were used to characterize Siglec-8 expression in sputum cells from stable asthma. An antibody-dependent cellular cytotoxicity (ADCC) assay was used to determine whether an S8mAb can decrease eosinophils in sputum from asthma patients ex vivo. A mast cell activation assay was used to determine whether an S8mAb can inhibit mast cell activation in human lung tissue ex vivo. RESULTS: Gene expression for Siglec-8 is increased in sputum cells in asthma and correlates with gene expression for eosinophils and mast cells. Gene expression for Siglec-8 is inversely and significantly correlated with measures of airflow obstruction in asthma patients. Siglec-8 is prominently expressed on the surface of eosinophils and mast cells in sputum. S8mAbs decrease eosinophils in sputum from patients with asthma and inhibit FcεR1-activated mast cells in lung tissues. CONCLUSIONS AND CLINICAL RELEVANCE: Siglec-8 is highly expressed on eosinophils and mast cells in asthmatic sputum and targeting Siglec-8 with an antibody is a plausible strategy to decrease sputum eosinophils and inhibit lung mast cells in asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Asthma/drug therapy , Eosinophils/drug effects , Lectins/antagonists & inhibitors , Lung/drug effects , Mast Cells/drug effects , Adult , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Apoptosis/drug effects , Asthma/immunology , Asthma/metabolism , Case-Control Studies , Cells, Cultured , Eosinophils/immunology , Eosinophils/metabolism , Female , Humans , Lectins/genetics , Lectins/immunology , Lectins/metabolism , Lung/immunology , Lung/metabolism , Male , Mast Cells/immunology , Mast Cells/metabolism , Middle Aged , Receptors, IgE/genetics , Receptors, IgE/metabolism , Sputum/cytology , Young Adult
14.
Biomaterials ; 214: 119225, 2019 09.
Article in English | MEDLINE | ID: mdl-31154151

ABSTRACT

The lymphatic system is an active player in the pathogenesis of several human diseases, including lymphedema and cancer. Relevant models are needed to advance our understanding of lymphatic biology in disease progression to improve therapy and patient outcomes. Currently, there are few 3D in vitro lymphatic models that can recapitulate the physiological structure, function, and interactions of lymphatic vessels in normal and diseased microenvironments. Here, we developed a 3D microscale lymphatic vessel (µLYMPH) system for generating human lymphatic vessels with physiological tubular structure and function. Consistent with characteristics of lymphatic vessels in vivo, the endothelium of cultured vessels was leaky with an average permeability of 1.38 × 10-5 ± 0.29 × 10-5 cm/s as compared to 0.68 × 10-5 ± 0.13 × 10-5 cm/s for blood vessels. This leakiness also resulted in higher uptake of solute by the lymphatic vessels under interstitial flow, demonstrating recapitulation of their natural draining function. The vessels secreted appropriate growth factors and inflammatory mediators. Our system identified the follistatin/activin axis as a novel pathway in lymphatic vessel maintenance and inflammation. Moreover, the µLYMPH system provided a platform for examining crosstalk between lymphatic vessels and tumor microenvironmental components, such as breast cancer-associated fibroblasts (CAFs). In co-culture with CAFs, vessel barrier function was significantly impaired by CAF-secreted IL-6, a possible pro-metastatic mechanism of lymphatic metastasis. Targeted blocking of the IL-6/IL-6R signaling pathway with an IL-6 neutralizing antibody fully rescued the vessels, demonstrating the potential of our system for screening therapeutic targets. These results collectively demonstrate the µLYMPH system as a powerful model for advancing lymphatic biology in health and disease.


Subject(s)
Lymphatic Vessels/physiology , Organ Culture Techniques/instrumentation , Cell Line , Cellular Microenvironment , Endothelial Cells/cytology , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lymphangiogenesis , Lymphatic Vessels/cytology , Permeability , Signal Transduction
16.
PLoS Pathog ; 12(4): e1005555, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27058347

ABSTRACT

The immune mechanisms that recognize inhaled Aspergillus fumigatus conidia to promote their elimination from the lungs are incompletely understood. FleA is a lectin expressed by Aspergillus fumigatus that has twelve binding sites for fucosylated structures that are abundant in the glycan coats of multiple plant and animal proteins. The role of FleA is unknown: it could bind fucose in decomposed plant matter to allow Aspergillus fumigatus to thrive in soil, or it may be a virulence factor that binds fucose in lung glycoproteins to cause Aspergillus fumigatus pneumonia. Our studies show that FleA protein and Aspergillus fumigatus conidia bind avidly to purified lung mucin glycoproteins in a fucose-dependent manner. In addition, FleA binds strongly to macrophage cell surface proteins, and macrophages bind and phagocytose fleA-deficient (∆fleA) conidia much less efficiently than wild type (WT) conidia. Furthermore, a potent fucopyranoside glycomimetic inhibitor of FleA inhibits binding and phagocytosis of WT conidia by macrophages, confirming the specific role of fucose binding in macrophage recognition of WT conidia. Finally, mice infected with ΔfleA conidia had more severe pneumonia and invasive aspergillosis than mice infected with WT conidia. These findings demonstrate that FleA is not a virulence factor for Aspergillus fumigatus. Instead, host recognition of FleA is a critical step in mechanisms of mucin binding, mucociliary clearance, and macrophage killing that prevent Aspergillus fumigatus pneumonia.


Subject(s)
Aspergillus fumigatus/immunology , Lectins/immunology , Macrophages/immunology , Mucins/immunology , Pulmonary Aspergillosis/immunology , Adult , Animals , Aspergillus fumigatus/pathogenicity , Blotting, Western , Disease Models, Animal , Female , Flow Cytometry , Fluorescent Antibody Technique , Fucose/metabolism , Fungal Proteins/immunology , Fungal Proteins/metabolism , Humans , Immunity, Mucosal/immunology , Lectins/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mucins/metabolism , Pulmonary Aspergillosis/metabolism , Spores, Fungal/immunology
17.
Sci Transl Med ; 7(276): 276ra27, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25717100

ABSTRACT

Airway mucus in cystic fibrosis (CF) is highly elastic, but the mechanism behind this pathology is unclear. We hypothesized that the biophysical properties of CF mucus are altered because of neutrophilic oxidative stress. Using confocal imaging, rheology, and biochemical measures of inflammation and oxidation, we found that CF airway mucus gels have a molecular architecture characterized by a core of mucin covered by a web of DNA and a rheological profile characterized by high elasticity that can be normalized by chemical reduction. We also found that high levels of reactive oxygen species in CF mucus correlated positively and significantly with high concentrations of the oxidized products of cysteine (disulfide cross-links). To directly determine whether oxidation can cross-link mucins to increase mucus elasticity, we exposed induced sputum from healthy subjects to oxidizing stimuli and found a marked and thiol-dependent increase in sputum elasticity. Targeting mucin disulfide cross-links using current thiol-amino structures such as N-acetylcysteine (NAC) requires high drug concentrations to have mucolytic effects. We therefore synthesized a thiol-carbohydrate structure (methyl 6-thio-6-deoxy-α-D-galactopyranoside) and found that it had stronger reducing activity than NAC and more potent and fast-acting mucolytic activity in CF sputum. Thus, oxidation arising from airway inflammation or environmental exposure contributes to pathologic mucus gel formation in the lung, which suggests that it can be targeted by thiol-modified carbohydrates.


Subject(s)
Cross-Linking Reagents/metabolism , Gels/metabolism , Lung/physiology , Mucins/metabolism , Mucus/metabolism , Polymers/metabolism , Acetylcysteine/pharmacology , Animals , Biomechanical Phenomena/drug effects , Cystic Fibrosis/pathology , Cystic Fibrosis/physiopathology , DNA/metabolism , Disulfides/metabolism , Elasticity/drug effects , Expectorants/pharmacology , Galactose/chemistry , Galactose/pharmacology , Humans , Lung/drug effects , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reducing Agents/pharmacology , Sputum/drug effects , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology
19.
PLoS One ; 8(12): e83077, 2013.
Article in English | MEDLINE | ID: mdl-24340081

ABSTRACT

Aspergillus fumigatus is an important allergen and opportunistic pathogen. Similarly to many other pathogens, it is able to produce lectins that may be involved in the host-pathogen interaction. We focused on the lectin AFL, which was prepared in recombinant form and characterized. Its binding properties were studied using hemagglutination and glycan array analysis. We determined the specificity of the lectin towards l-fucose and fucosylated oligosaccharides, including α1-6 linked core-fucose, which is an important marker for cancerogenesis. Other biologically relevant saccharides such as sialic acid, d-mannose or d-galactose were not bound. Blood group epitopes of the ABH and Lewis systems were recognized, Le(Y) being the preferred ligand among others. To provide a correlation between the observed functional characteristics and structural basis, AFL was crystallized in a complex with methyl-α,L-selenofucoside and its structure was solved using the SAD method. Six binding sites, each with different compositions, were identified per monomer and significant differences from the homologous AAL lectin were found. Structure-derived peptides were utilized to prepare anti-AFL polyclonal antibodies, which suggested the presence of AFL on the Aspergillus' conidia, confirming its expression in vivo. Stimulation of human bronchial cells by AFL led to IL-8 production in a dose-dependent manner. AFL thus probably contributes to the inflammatory response observed upon the exposure of a patient to A. fumigatus. The combination of affinity to human epithelial epitopes, production by conidia and pro-inflammatory activity is remarkable and shows that AFL might be an important virulence factor involved in an early stage of A. fumigatus infection.


Subject(s)
Aspergillus fumigatus/chemistry , Fucose/chemistry , Lectins/chemistry , Spores, Fungal/chemistry , Amino Acid Sequence , Aspergillosis/immunology , Binding Sites , Bronchi/cytology , Bronchi/microbiology , Epitopes/chemistry , Galactose/chemistry , Genome, Fungal , Hemagglutination , Host-Pathogen Interactions , Humans , Interleukin-8/metabolism , Mannose/chemistry , Molecular Sequence Data , N-Acetylneuraminic Acid/chemistry , Oligosaccharides/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Virulence Factors/chemistry
20.
J Org Chem ; 76(2): 724-7, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21190367

ABSTRACT

Original glycodendrimers emanating from propargylated hexaphenylbenzene cores and containing up to 54 peripheral sugar ligands have been synthesized by Cu(I)-catalyzed [1,3]-dipolar cycloadditions using both convergent and divergent approaches.


Subject(s)
Carbohydrates/chemical synthesis , Glycoconjugates/chemistry , Glycoconjugates/chemical synthesis , Macromolecular Substances/chemistry , Pargyline/chemistry , Carbohydrates/chemistry , Catalysis , Copper/chemistry , Glycosylation , Ligands , Models, Molecular , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...