Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 579: 216468, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37940068

ABSTRACT

Bone metastatic disease of prostate cancer (PCa) is incurable and progression in bone is largely dictated by tumor-stromal interactions in the bone microenvironment. We showed previously that bone neutrophils initially inhibit bone metastatic PCa growth yet metastatic PCa becomes resistant to neutrophil response. Further, neutrophils isolated from tumor-bone lost their ability to suppress tumor growth through unknown mechanisms. With this study, our goal was to define the impact of metastatic PCa on neutrophil function throughout tumor progression and to determine the potential of neutrophils as predictive biomarkers of metastatic disease. Using patient peripheral blood polymorphonuclear neutrophils (PMNs), we identified that PCa progression dictates PMN cell surface markers and gene expression, but not cytotoxicity against PCa. Importantly, we also identified a novel phenomenon in which second generation androgen deprivation therapy (ADT) suppresses PMN cytotoxicity via increased transforming growth factor beta receptor I (TßRI). High dose testosterone and genetic or pharmacologic TßRI inhibition rescued androgen receptor-mediated neutrophil suppression and restored neutrophil anti-tumor immune response. These studies highlight the ability to leverage standard-care ADT to generate neutrophil anti-tumor responses against bone metastatic PCa.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgens , Neutrophils/metabolism , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Cell Line, Tumor , Tumor Microenvironment
2.
Cancer Immunol Immunother ; 69(6): 1113-1130, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32114681

ABSTRACT

Bone metastatic prostate cancer (BM-PCa) significantly reduces overall patient survival and is currently incurable. Current standard immunotherapy showed promising results for PCa patients with metastatic, but less advanced, disease (i.e., fewer than 20 bone lesions) suggesting that PCa growth in bone contributes to response to immunotherapy. We found that: (1) PCa stimulates recruitment of neutrophils, the most abundant immune cell in bone, and (2) that neutrophils heavily infiltrate regions of prostate tumor in bone of BM-PCa patients. Based on these findings, we examined the impact of direct neutrophil-prostate cancer interactions on prostate cancer growth. Bone marrow neutrophils directly induced apoptosis of PCa in vitro and in vivo, such that neutrophil depletion in bone metastasis models enhanced BM-PCa growth. Neutrophil-mediated PCa killing was found to be mediated by suppression of STAT5, a transcription factor shown to promote PCa progression. However, as the tumor progressed in bone over time, neutrophils from late-stage bone tumors failed to elicit cytotoxic effector responses to PCa. These findings are the first to demonstrate that bone-resident neutrophils inhibit PCa and that BM-PCa are able to progress via evasion of neutrophil-mediated killing. Enhancing neutrophil cytotoxicity in bone may present a novel therapeutic option for bone metastatic prostate cancer.


Subject(s)
Bone Neoplasms/secondary , Neutrophils/metabolism , Prostatic Neoplasms/blood , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Neutrophils/cytology , Prostatic Neoplasms/complications , Prostatic Neoplasms/pathology
3.
Trends Cancer ; 5(12): 789-798, 2019 12.
Article in English | MEDLINE | ID: mdl-31813456

ABSTRACT

Immunotherapy has emerged at the forefront of cancer therapy; however, patient survival remains low for many cancer types. In consideration of this, non-T cell immune populations, such as innate immune cells, have been identified as potential immunotherapeutic targets. In noncancerous settings, neutrophils are first responders to injury and infection, and work in a partnership with macrophages to regulate inflammation. However, the diversity of tumor-associated neutrophils (TANs) remains elusive. Furthermore, it is likely that TANs and tumor-associated macrophages (TAMs) act in tandem within tumors and contribute both contrasting and synergistic roles in tumor progression. In this Opinion, we discuss the complexity of TAN and TAM functions, the interplay between TANs and TAMs, and major considerations required for implementing TAN/TAM-based therapies.


Subject(s)
Immunity, Innate/immunology , Immunotherapy/methods , Macrophages/metabolism , Neutrophils/metabolism , Tumor Microenvironment/immunology , Humans
4.
Cancers (Basel) ; 11(9)2019 Aug 24.
Article in English | MEDLINE | ID: mdl-31450600

ABSTRACT

Fucosylation is a post-translational modification of glycans, proteins, and lipids that is responsible for many biological processes. Fucose conjugation via α(1,2), α(1,3), α(1,4), α(1,6), and O'- linkages to glycans, and variations in fucosylation linkages, has important implications for cancer biology. This review focuses on the roles that fucosylation plays in cancer, specifically through modulation of cell surface proteins and signaling pathways. How L-fucose and serum fucosylation patterns might be used for future clinical diagnostic, prognostic, and therapeutic approaches will be discussed.

5.
Physiol Rep ; 7(7): e14034, 2019 04.
Article in English | MEDLINE | ID: mdl-30972920

ABSTRACT

Overdevelopment of visceral adipose is positively correlated with the etiology of obesity-associated pathologies including cardiovascular disease and insulin resistance. However, identification of genetic, molecular, and physiological factors regulating adipose development and function in response to nutritional stress is incomplete. Fibroblast Growth Factor 1 (FGF1) is a cytokine expressed and released by both adipocytes and endothelial cells under hypoxia, thermal, and oxidative stress. Expression of Fibroblast Growth Factor 1 (FGF1) in adipose is required for normal depot development and remodeling. Loss of FGF1 leads to deleterious changes in adipose morphology, metabolism, and insulin resistance. Conversely, diabetic and obese mice injected with recombinant FGF1 display improvements in insulin sensitivity and a reduction in adiposity. We report in this novel, in vivo study that transgenic mice expressing an endothelial-specific FGF1 transgene (FGF1-Tek) are resistant to high-fat diet-induced abdominal adipose accretion and are more glucose-tolerant than wild-type control animals. Metabolic chamber analyses indicate that suppression of the development of visceral adiposity and insulin resistance was not associated with alterations in appetite or resting metabolic rate in the FGF1-Tek strain. Instead, FGF1-Tek mice display increased locomotor activity that likely promotes the utilization of dietary fatty acids before they can accumulate in adipose and liver. This study provides insight into the impact that genetic differences dictating the production of FGF1 has on the risk for developing obesity-related metabolic disease in response to nutritional stress.


Subject(s)
Adipose Tissue/metabolism , Endothelial Cells/metabolism , Fibroblast Growth Factor 1/genetics , Locomotion/genetics , Obesity, Abdominal/genetics , Adipocytes/metabolism , Adiposity/drug effects , Adiposity/genetics , Animals , Blood Glucose/metabolism , Diet, High-Fat , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 1/pharmacology , Insulin/blood , Insulin Resistance/genetics , Liver/metabolism , Mice , Mice, Transgenic , Obesity, Abdominal/metabolism
6.
PLoS One ; 13(6): e0199128, 2018.
Article in English | MEDLINE | ID: mdl-29924834

ABSTRACT

The fucose salvage pathway is a two-step process in which mammalian cells transform L-fucose into GDP-L-fucose, a universal fucose donor used by fucosyltransferases to modify glycans. Emerging evidence indicates the fucose salvage pathway and the fucosylation of proteins are altered during melanoma progression and metastasis. However the underlying mechanisms are not completely understood. Here, we report that the fucose salvage pathway inhibits invadopodia formation and extracellular matrix degradation by promoting α-1,2 fucosylation. Chemically or genetically increasing the fucose salvage pathway decreases invadopodium numbers and inhibits the proteolytic activity of invadopodia in WM793 melanoma cells. Inhibiting fucosylation by depleting fucokinase abrogates L-fucose-mediated inhibition of invadopodia, suggesting dependence on the fucose salvage pathway. The inhibition of invadopodium formation by L-fucose or ectopically expressed FUK could be rescued by treatment with α-1,2, but not α-1,3/α-1,4 fucosidase, implicating an α-1,2 fucose linkage-dependent anti-metastatic effect. The expression of FUT1, an α-1,2 fucosyltransferase, is remarkably down-regulated during melanoma progression, and the ectopic expression of FUT1 is sufficient to inhibit invadopodium formation and ECM degradation. Our findings indicate that the fucose salvage pathway can inhibit invadopodium formation, and consequently, invasiveness in melanoma via α-1,2 fucosylation. Re-activation of this pathway in melanoma could be useful for preventing melanoma invasion and metastasis.


Subject(s)
Extracellular Matrix/metabolism , Fucose/metabolism , Fucosyltransferases/physiology , Melanoma/metabolism , Neoplasm Proteins/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Podosomes/physiology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Fucose/pharmacology , Fucosyltransferases/deficiency , Fucosyltransferases/genetics , Genetic Vectors/pharmacology , Glycosylation , Humans , Melanoma/physiopathology , Metabolic Networks and Pathways , Neoplasm Invasiveness , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Podosomes/drug effects , Protein Processing, Post-Translational , Recombinant Proteins/pharmacology , Up-Regulation , Galactoside 2-alpha-L-fucosyltransferase
7.
J Biol Chem ; 291(53): 27323-27333, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27879315

ABSTRACT

Fascin is an actin bundling protein that cross-links individual actin filaments into straight, compact, and stiff bundles, which are crucial for the formation of filopodia, stereocillia, and other finger-like membrane protrusions. The dysregulation of fascin has been implicated in cancer metastasis, hearing loss, and blindness. Here we identified monoubiquitination as a novel mechanism that regulates fascin bundling activity and dynamics. The monoubiquitination sites were identified to be Lys247 and Lys250, two residues located in a positive charge patch at the actin binding site 2 of fascin. Using a chemical ubiquitination method, we synthesized chemically monoubiquitinated fascin and determined the effects of monoubiquitination on fascin bundling activity and dynamics. Our data demonstrated that monoubiquitination decreased the fascin bundling EC50, delayed the initiation of bundle assembly, and accelerated the disassembly of existing bundles. By analyzing the electrostatic properties on the solvent-accessible surface of fascin, we proposed that monoubiquitination introduced steric hindrance to interfere with the interaction between actin filaments and the positively charged patch at actin binding site 2. We also identified Smurf1 as a E3 ligase regulating the monoubiquitination of fascin. Our findings revealed a previously unidentified regulatory mechanism for fascin, which will have important implications for the understanding of actin bundle regulation under physiological and pathological conditions.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Microfilament Proteins/metabolism , Ubiquitin/metabolism , Animals , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Rats , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
8.
Mol Cell Oncol ; 2(4)2015.
Article in English | MEDLINE | ID: mdl-26346228

ABSTRACT

Dysregulated calcium signaling has been increasingly implicated in tumor dissemination and progression. In a recent study we investigated the mechanism underlying calcium-mediated melanoma invasion and metastasis, and discovered that hyperactive Ca2+ oscillation in melanoma cells promoted invasion and metastasis through promoting invadopodium formation and extracellular matrix remodeling.

SELECTION OF CITATIONS
SEARCH DETAIL
...