Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 591: 216879, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38636895

ABSTRACT

Galectin-3 (Gal-3) is a multifunctional protein that plays a pivotal role in the initiation and progression of various central nervous system diseases, including cancer. Although the involvement of Gal-3 in tumour progression, resistance to treatment and immunosuppression has long been studied in different cancer types, mainly outside the central nervous system, its elevated expression in myeloid and glial cells underscores its profound impact on the brain's immune response. In this context, microglia and infiltrating macrophages, the predominant non-cancerous cells within the tumour microenvironment, play critical roles in establishing an immunosuppressive milieu in diverse brain tumours. Through the utilisation of primary cell cultures and immortalised microglial cell lines, we have elucidated the central role of Gal-3 in promoting cancer cell migration, invasion, and an immunosuppressive microglial phenotypic activation. Furthermore, employing two distinct in vivo models encompassing primary (glioblastoma) and secondary brain tumours (breast cancer brain metastasis), our histological and transcriptomic analysis show that Gal-3 depletion triggers a robust pro-inflammatory response within the tumour microenvironment, notably based on interferon-related pathways. Interestingly, this response is prominently observed in tumour-associated microglia and macrophages (TAMs), resulting in the suppression of cancer cells growth.


Subject(s)
Brain Neoplasms , Cell Movement , Cell Proliferation , Galectin 3 , Glioblastoma , Microglia , Tumor Microenvironment , Microglia/metabolism , Microglia/pathology , Galectin 3/metabolism , Galectin 3/genetics , Humans , Animals , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Glioblastoma/immunology , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Line, Tumor , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Macrophages/metabolism , Macrophages/immunology , Neoplasm Invasiveness , Blood Proteins/metabolism , Galectins/metabolism , Galectins/genetics , Signal Transduction , Mice , Gene Expression Regulation, Neoplastic
2.
J Vis Exp ; (196)2023 06 02.
Article in English | MEDLINE | ID: mdl-37335107

ABSTRACT

One of the biggest challenges in developing effective therapies against glioblastoma is overcoming the strong immune suppression within the tumor microenvironment. Immunotherapy has emerged as an effective strategy to turn the immune system response against tumor cells. Glioma-associated macrophages and microglia (GAMs) are major drivers of such anti-inflammatory scenarios. Therefore, enhancing the anti-cancerous response in GAMs may represent a potential co-adjuvant therapy to treat glioblastoma patients. In that vein, fungal ß-glucan molecules have long been known as potent immune modulators. Their ability to stimulate the innate immune activity and improve treatment response has been described. Those modulating features are partly attributed to their ability to bind to pattern recognition receptors, which, interestingly, are greatly expressed in GAMs. Thus, this work is focused on the isolation, purification, and subsequent use of fungal ß-glucans to enhance the tumoricidal response of microglia against glioblastoma cells. The mouse glioblastoma (GL261) and microglia (BV-2) cell lines are used to test the immunomodulatory properties of four different fungal ß-glucans extracted from mushrooms heavily used in the current biopharmaceutical industry: Pleurotus ostreatus, Pleurotus djamor, Hericium erinaceus, and Ganoderma lucidum. To test these compounds, co-stimulation assays were performed to measure the effect of a pre-activated microglia-conditioned medium on the proliferation and apoptosis activation in glioblastoma cells.


Subject(s)
Glioblastoma , Glioma , beta-Glucans , Animals , Mice , Glioblastoma/pathology , beta-Glucans/pharmacology , beta-Glucans/metabolism , Macrophages/metabolism , Glioma/pathology , Microglia/metabolism , Immunotherapy , Tumor Microenvironment
3.
Cancer Res ; 80(24): 5642-5655, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33106335

ABSTRACT

Astrocytes are thought to play a pivotal role in coupling neural activity and cerebral blood flow. However, it has been shown that astrocytes undergo morphologic changes in response to brain metastasis, switching to a reactive phenotype, which has the potential to significantly compromise cerebrovascular function and contribute to the neurological sequelae associated with brain metastasis. Given that STAT3 is a key regulator of astrocyte reactivity, we aimed here to determine the impact of STAT3-mediated astrocyte reactivity on neurovascular function in brain metastasis. Rat models of brain metastasis and ciliary neurotrophic factor were used to induce astrocyte reactivity. Multimodal imaging, electrophysiology, and IHC were performed to determine the relationship between reactive astrocytes and changes in the cerebrovascular response to electrical and physiological stimuli. Subsequently, the STAT3 pathway in astrocytes was inhibited with WP1066 to determine the role of STAT3-mediated astrocyte reactivity, specifically, in brain metastasis. Astrocyte reactivity associated with brain metastases impaired cerebrovascular responses to stimuli at both the cellular and functional level and disrupted astrocyte-endothelial interactions in both animal models and human brain metastasis samples. Inhibition of STAT3-mediated astrocyte reactivity in rats with brain metastases restored cerebrovascular function, as shown by in vivo imaging, and limited cerebrovascular changes associated with tumor growth. Together these findings suggest that inhibiting STAT3-mediated astrocyte reactivity may confer significant improvements in neurological outcome for patients with brain metastases and could potentially be tested in other brain tumors. SIGNIFICANCE: These findings demonstrate that selectively targeting STAT3-mediated astrocyte reactivity ameliorates the cerebrovascular dysfunction associated with brain metastasis, providing a potential therapeutic avenue for improved patient outcome.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Animals , Astrocytes/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Cerebrovascular Circulation , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Female , Humans , Laser Speckle Contrast Imaging , Magnetic Resonance Spectroscopy , Multimodal Imaging , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/pathology , Pyridines/pharmacology , Rats , Rats, Inbred Strains , Tyrphostins/pharmacology
4.
Mol Cancer Ther ; 18(11): 2030-2042, 2019 11.
Article in English | MEDLINE | ID: mdl-31395687

ABSTRACT

The role of Notch signaling and its ligand JAGGED1 (JAG1) in tumor biology has been firmly established, making them appealing therapeutic targets for cancer treatment. Here, we report the development and characterization of human/rat-specific JAG1-neutralizing mAbs. Epitope mapping identified their binding to the Notch receptor interaction site within the JAG1 Delta/Serrate/Lag2 domain, where E228D substitution prevented effective binding to the murine Jag1 ortholog. These antibodies were able to specifically inhibit JAG1-Notch binding in vitro, downregulate Notch signaling in cancer cells, and block the heterotypic JAG1-mediated Notch signaling between endothelial and vascular smooth muscle cells. Functionally, in vitro treatment impaired three-dimensional growth of breast cancer cell spheroids, in association with a reduction in cancer stem cell number. In vivo testing showed variable effects on human xenograft growth when only tumor-expressed JAG1 was targeted (mouse models) but a more robust effect when stromal-expressed Jag1 was also targeted (rat MDA-MB-231 xenograft model). Importantly, treatment of established triple receptor-negative breast cancer brain metastasis in rats showed a significant reduction in neoplastic growth. MRI imaging demonstrated that this was associated with a substantial improvement in blood-brain barrier function and tumor perfusion. Lastly, JAG1-targeting antibody treatment did not cause any detectable toxicity, further supporting its clinical potential for cancer therapy.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Jagged-1 Protein/chemistry , Jagged-1 Protein/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Development , Female , Humans , Mice , Rats , Receptors, Notch/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
5.
Nat Commun ; 8: 14254, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28198362

ABSTRACT

Ligand-conjugated microparticles of iron oxide (MPIO) have the potential to provide high sensitivity contrast for molecular magnetic resonance imaging (MRI). However, the accumulation and persistence of non-biodegradable micron-sized particles in liver and spleen precludes their clinical use and limits the translational potential of MPIO-based contrast agents. Here we show that ligand-targeted MPIO derived from multiple iron oxide nanoparticles may be coupled covalently through peptide linkers that are designed to be cleaved by intracellular macrophage proteases. The synthesized particles possess potential characteristics for targeted MRI contrast agents, including high relaxivity, unappreciable sedimentation, clearance from circulation and no overt toxicity. Importantly, we demonstrate that these particles are rapidly degraded both in vitro and in vivo, and that the targeted probes can be used for detection of inflammation in vivo using MRI. This approach provides a platform for molecular MRI contrast agents that is potentially more suitable for translation to humans.


Subject(s)
Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Peptide Hydrolases/metabolism , Animals , Antibodies/metabolism , Contrast Media/chemistry , Ferric Compounds/chemistry , Humans , Magnetite Nanoparticles/ultrastructure , Male , Mice , Particle Size , RAW 264.7 Cells , Vascular Cell Adhesion Molecule-1/metabolism
6.
Int J Cancer ; 134(4): 885-96, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-23913394

ABSTRACT

Metastasis to the brain results in significant impairment of brain function and poor patient survival. Currently, magnetic resonance imaging (MRI) is under-utilised in monitoring brain metastases and their effects on brain function. Here, we sought to establish a model of focal brain metastasis in the rat that enables serial multimodal structural and functional MRI studies, and to assess the sensitivity of these approaches to metastatic growth. Female Berlin-Druckrey-IX rats were injected intracerebrally with metastatic ENU1564 cells in the ventroposterior medial nucleus (VPM) of the thalamus, a relay node of the whisker-to-barrel cortex pathway. Animals underwent multimodal structural and vascular MRI, as well as functional MRI of the cortical blood oxygenation level dependent (BOLD) responses to whisker pad stimulation. T2 , diffusion, magnetisation transfer and perfusion weighted MRI enabled differentiation between a central area of more advanced metastatic growth and penumbral regions of co-optive perivascular micrometastatic growth, with magnetisation transfer MRI being the most sensitive to micrometastatic growth. Areas of cortical BOLD activation in response to whisker pad stimulation were significantly reduced in the hemisphere containing metastases in the VPM. The reduction in BOLD response correlated with metastatic burden in the thalamus, and was sensitive to the presence of smaller metastases than currently detectable clinically. Our findings suggest that multimodal MRI provides greater sensitivity to tumour heterogeneity and micrometastatic growth than single modality contrast-enhanced MRI. Understanding the relationships between these MRI parameters and the underlying pathology may greatly enhance the utility of MRI in diagnosis, staging and monitoring of brain metastasis.


Subject(s)
Brain Neoplasms/secondary , Brain/pathology , Diffusion Magnetic Resonance Imaging , Magnetic Resonance Imaging , Mammary Neoplasms, Experimental/pathology , Multimodal Imaging , Animals , Biomarkers, Tumor/analysis , Brain/metabolism , Brain Neoplasms/metabolism , Female , Immunoenzyme Techniques , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Micrometastasis , Rats , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...