Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 80(7): 179, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37314567

ABSTRACT

Glioblastoma (GBM) is the most common and fatal primary tumor of the central nervous system (CNS) and current treatments have limited success. Chemokine signaling regulates both malignant cells and stromal cells of the tumor microenvironment (TME), constituting a potential therapeutic target against brain cancers. Here, we investigated the C-C chemokine receptor type 7 (CCR7) and the chemokine (C-C-motif) ligand 21 (CCL21) for their expression and function in human GBM and then assessed their therapeutic potential in preclinical mouse GBM models. In GBM patients, CCR7 expression positively associated with a poor survival. CCL21-CCR7 signaling was shown to regulate tumor cell migration and proliferation while also controlling tumor associated microglia/macrophage recruitment and VEGF-A production, thereby controlling vascular dysmorphia. Inhibition of CCL21-CCR7 signaling led to an increased sensitivity to temozolomide-induced tumor cell death. Collectively, our data indicate that drug targeting of CCL21-CCR7 signaling in tumor and TME cells is a therapeutic option against GBM.


Subject(s)
Glioblastoma , Microglia , Animals , Mice , Humans , Glioblastoma/drug therapy , Receptors, CCR7/genetics , Macrophages , Central Nervous System , Tumor Microenvironment , Chemokine CCL21
2.
J Mol Med (Berl) ; 100(10): 1405-1425, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36056255

ABSTRACT

Cellular prion protein (PrPC) is a highly conserved glycoprotein, present both anchored in the cell membrane and soluble in the extracellular medium. It has a diversity of ligands and is variably expressed in numerous tissues and cell subtypes, most notably in the central nervous system (CNS). Its importance has been brought to light over the years both under physiological conditions, such as embryogenesis and immune system homeostasis, and in pathologies, such as cancer and neurodegenerative diseases. During development, PrPC plays an important role in CNS, participating in axonal growth and guidance and differentiation of glial cells, but also in other organs such as the heart, lung, and digestive system. In diseases, PrPC has been related to several types of tumors, modulating cancer stem cells, enhancing malignant properties, and inducing drug resistance. Also, in non-neoplastic diseases, such as Alzheimer's and Parkinson's diseases, PrPC seems to alter the dynamics of neurotoxic aggregate formation and, consequently, the progression of the disease. In this review, we explore in detail the multiple functions of this protein, which proved to be relevant for understanding the dynamics of organism homeostasis, as well as a promising target in the treatment of both neoplastic and degenerative diseases.


Subject(s)
Neoplasms , Neurodegenerative Diseases , PrPC Proteins , Central Nervous System/metabolism , Humans , Neoplastic Stem Cells/metabolism , PrPC Proteins/genetics , PrPC Proteins/metabolism
3.
Signal Transduct Target Ther ; 6(1): 45, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33526777

ABSTRACT

Lysophosphatidic acid (LPA) is an abundant bioactive phospholipid, with multiple functions both in development and in pathological conditions. Here, we review the literature about the differential signaling of LPA through its specific receptors, which makes this lipid a versatile signaling molecule. This differential signaling is important for understanding how this molecule can have such diverse effects during central nervous system development and angiogenesis; and also, how it can act as a powerful mediator of pathological conditions, such as neuropathic pain, neurodegenerative diseases, and cancer progression. Ultimately, we review the preclinical and clinical uses of Autotaxin, LPA, and its receptors as therapeutic targets, approaching the most recent data of promising molecules modulating both LPA production and signaling. This review aims to summarize the most update knowledge about the mechanisms of LPA production and signaling in order to understand its biological functions in the central nervous system both in health and disease.


Subject(s)
Lysophospholipids/genetics , Neovascularization, Pathologic/genetics , Phospholipids/genetics , Humans , Lysophospholipids/metabolism , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Phospholipids/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/therapeutic use , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/therapeutic use , Signal Transduction/genetics
4.
Cytokine Growth Factor Rev ; 57: 73-84, 2021 02.
Article in English | MEDLINE | ID: mdl-32561134

ABSTRACT

Stress inducible protein 1 (STI1) is a co-chaperone acting with Hsp70 and Hsp90 for the correct client proteins' folding and therefore for the maintenance of cellular homeostasis. Besides being expressed in the cytosol, STI1 can also be found both in the cell membrane and the extracellular medium playing several relevant roles in the central nervous system (CNS) and tumor microenvironment. During CNS development, in association with cellular prion protein (PrPc), STI1 regulates crucial events such as neuroprotection, neuritogenesis, astrocyte differentiation and survival. In cancer, STI1 is involved with tumor growth and invasion, is undoubtedly a pro-tumor factor, being considered as a biomarker and possibly therapeutic target for several malignancies. In this review, we discuss current knowledge and new findings on STI1 function as well as its role in tissue homeostasis, CNS and tumor progression.


Subject(s)
Molecular Chaperones , Heat-Shock Proteins , Humans , Tumor Microenvironment
5.
Trends Cancer ; 5(1): 46-65, 2019 01.
Article in English | MEDLINE | ID: mdl-30616755

ABSTRACT

Glioblastoma (GBM) is the most common and fatal primary malignant brain tumor. Despite advances in the understanding of the biology of gliomas, little has changed in the treatment of these tumors in the past decade. Phase III clinical trials showed no benefit for the use of bevacizumab in newly diagnosed patients, leading to a renewed search for new antiangiogenic drugs, as well as immunotherapeutic approaches, including checkpoint inhibitors, chimeric antigen receptor T cells, and intracerebral CpG-oligodeoxynucleotides. The emerging role of infiltrating microglia and macrophages, and of metabolic alterations, is also being taken into account in preclinical research and drug development. In this review, we discuss progress in the search for new therapeutic strategies, particularly approaches focusing on the tumor microenvironment.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Molecular Targeted Therapy , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Brain Neoplasms/etiology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Energy Metabolism/drug effects , Genetic Therapy , Glioblastoma/etiology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunotherapy, Adoptive/methods , Molecular Targeted Therapy/methods , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
6.
Mol Neurobiol ; 56(2): 1517-1530, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29948952

ABSTRACT

Glioblastoma is an extremely aggressive and deadly brain tumor known for its striking cellular heterogeneity and capability to communicate with microenvironment components, such as microglia. Microglia-glioblastoma interaction contributes to an increase in tumor invasiveness, and Wnt signaling pathway is one of the main cascades related to tumor progression through changes in cell migration and invasion. However, very little is known about the role of canonical Wnt signaling during microglia-glioblastoma crosstalk. Here, we show for the first time that Wnt3a is one of the factors that regulate interactions between microglia and glioblastoma cells. Wnt3a activates the Wnt/ß-catenin signaling of both glioblastoma and microglial cells. Glioblastoma-conditioned medium not only induces nuclear translocation of microglial ß-catenin but also increases microglia viability and proliferation as well as Wnt3a, cyclin-D1, and c-myc expression. Moreover, glioblastoma-derived Wnt3a increases microglial ARG-1 and STI1 expression, followed by an upregulation of IL-10 mRNA levels, and a decrease in IL1ß gene expression. The presence of Wnt3a in microglia-glioblastoma co-cultures increases the formation of membrane nanotubes accompanied by changes in migration capability. In vivo, tumors formed from Wnt3a-stimulated glioblastoma cells presented greater microglial infiltration and more aggressive characteristics such as growth rate than untreated tumors. Thus, we propose that Wnt3a belongs to the arsenal of factors capable of stimulating the induction of M2-like phenotype on microglial cells, which contributes to the poor prognostic of glioblastoma, reinforcing that Wnt/ß-catenin pathway can be a potential therapeutic target to attenuate glioblastoma progression.


Subject(s)
Microglia/metabolism , Wnt Signaling Pathway/physiology , Wnt3A Protein/metabolism , beta Catenin/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Glioblastoma/genetics , Humans , Phenotype
7.
Mol Neurobiol ; 55(11): 8668-8679, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29582399

ABSTRACT

Perinatal asphyxia remains a significant cause of neonatal mortality and is associated with long-term neurodegenerative disorders. In the present study, we evaluated cellular and subcellular damages to brain development in a model of mild perinatal asphyxia. Survival rate in the experimental group was 67%. One hour after the insult, intraperitoneally injected Evans blue could be detected in the fetuses' brains, indicating disruption of the blood-brain barrier. Although brain mass and absolute cell numbers (neurons and non-neurons) were not reduced after perinatal asphyxia immediately and in late brain development, subcellular alterations were detected. Cortical oxygen consumption increased immediately after asphyxia, and remained high up to 7 days, returning to normal levels after 14 days. We observed an increased resistance to mitochondrial membrane permeability transition, and calcium buffering capacity in asphyxiated animals from birth to 14 days after the insult. In contrast to ex vivo data, mitochondrial oxygen consumption in primary cell cultures of neurons and astrocytes was not altered after 1% hypoxia. Taken together, our results demonstrate that although newborns were viable and apparently healthy, brain development is subcellularly altered by perinatal asphyxia. Our findings place the neonate brain mitochondria as a potential target for therapeutic protective interventions.


Subject(s)
Asphyxia/pathology , Brain/growth & development , Brain/pathology , Mitochondria/pathology , Animals , Animals, Newborn , Asphyxia/blood , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Cell Hypoxia , Cell Respiration , Cells, Cultured , Citrate (si)-Synthase/metabolism , Energy Metabolism , Female , Lactates/blood , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Neurons/metabolism , Neurons/pathology , Organ Size , Permeability , Rats, Wistar , Survival Analysis
8.
Mol Neurobiol ; 55(8): 6816-6833, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29349577

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor and still lacks effective therapeutic strategies. It has already been shown that old drugs like sulfasalazine (SAS) and valproic acid (VPA) present antitumoral activities in glioma cell lines. SAS has also been associated with a decrease of intracellular glutathione (GSH) levels through a potent inhibition of xc- glutamate/cystine exchanger leading to an antioxidant deprotection. In the same way, VPA was recently identified as a histone deacetylase (HDAT) inhibitor capable of activating tumor suppression genes. As both drugs are widely used in clinical practice and their profile of adverse effects is well known, the aim of our study was to investigate the effects of the combined treatment with SAS and VPA in GBM cell lines. We observed that both drugs were able to reduce cell viability in a dose-dependent manner and the combined treatment potentiated these effects. Combined treatment also increased cell death and inhibited proliferation of GBM cells, while having no effect on human and rat cultured astrocytes. Also, we observed high protein expression of the catalytic subunit of xc- in all the examined GBM cell lines, and treatment with SAS blocked its activity and decreased intracellular GSH levels. Noteworthy, SAS but not VPA was also able to reduce the [14C]-ascorbate uptake. Together, these data indicate that SAS and VPA exhibit a substantial effect on GBM cell's death related to an intracellular oxidative response imbalance, making this combination of drugs a promising therapeutic strategy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Intracellular Space/metabolism , Sulfasalazine/pharmacology , Valproic Acid/pharmacology , Amino Acid Transport System y+/metabolism , Animals , Ascorbic Acid/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Therapy, Combination , Glutathione/metabolism , Humans , Mesoderm/drug effects , Mesoderm/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Oxidation-Reduction , Rats , Time Factors
9.
Adv Exp Med Biol ; 949: 245-261, 2016.
Article in English | MEDLINE | ID: mdl-27714693

ABSTRACT

Glioblastoma is a malignant tumor of astrocytic origin that is highly invasive, proliferative and angiogenic. Despite current advances in multimodal therapies, such as surgery, radio- and chemotherapy, the outcome for patients with glioblastoma is nearly always fatal. The glioblastoma microenvironment has a tremendous influence over the tumor growth and spread. Microglia and macrophages are abundant cells in the tumor mass. Increasing evidence indicates that glioblastoma recruits these cell populations and signals in a way that microglia and macrophages are subverted to promote tumor progression. In this chapter, we discuss some aspects of the interaction between microglia and glioblastoma, consequences of this interaction for tumor progression and the possibility of microglial cells being used as therapeutic vectors, which opens up new alternatives for the development of GBM therapies targeting microglia.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Macrophages/metabolism , Microglia/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Communication/drug effects , Cell Communication/radiation effects , Cytokines/genetics , Cytokines/metabolism , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gamma Rays/therapeutic use , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Macrophages/pathology , Microglia/pathology , Oligodeoxyribonucleotides/therapeutic use , Signal Transduction , Temozolomide , Treatment Failure , Tumor Microenvironment
10.
Cancer Cell Int ; 16: 46, 2016.
Article in English | MEDLINE | ID: mdl-27330409

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common primary brain tumor presenting self-renewing cancer stem cells. The role of these cells on the development of the tumors has been proposed to recapitulate programs from embryogenesis. Recently, the embryonic transforming growth factor-ß (TGF-ß) protein Nodal has been shown to be reactivated upon tumor development; however, its availability in GBM cells has not been addressed so far. In this study, we investigated by an original approach the mechanisms that dynamically control both intra and extracellular Nodal availability during GBM tumorigenesis. METHODS: We characterized the dynamics of Nodal availability in both stem and more differentiated GBM cells through morphological analysis, immunofluorescence of Nodal protein and of early (EEA1 and Rab5) and late (Rab7 and Rab11) endocytic markers and Western Blot. Tukey's test was used to analyze the prevalent correlation of Nodal with different endocytic markers inside specific differentiation states, and Sidak's multiple comparisons test was used to compare the prevalence of Nodal/endocytic markers co-localization between two differentiation states of GBM cells. Paired t test was used to analyze the abundance of Nodal protein, in extra and intracellular media. RESULTS: The cytoplasmic distribution of Nodal was dynamically regulated and strongly correlated with the differentiation status of GBM cells. While Nodal-positive vesicle-like particles were symmetrically distributed in GBM stem cells (GBMsc), they presented asymmetric perinuclear localization in more differentiated GBM cells (mdGBM). Strikingly, when subjected to dedifferentiation, the distribution of Nodal in mdGBM shifted to a symmetric pattern. Moreover, the availability of both intracellular and secreted Nodal were downregulated upon GBMsc differentiation, with cells becoming elongated, negative for Nodal and positive for Nestin. Interestingly, the co-localization of Nodal with endosomal vesicles also depended on the differentiation status of the cells, with Nodal seen more packed in EEA1/Rab5 + vesicles in GBMsc and more in Rab7/11 + vesicles in mdGBM. CONCLUSIONS: Our results show for the first time that Nodal availability relates to GBM cell differentiation status and that it is dynamically regulated by an endocytic pathway during GBM tumorigenesis, shedding new light on molecular pathways that might emerge as putative targets for Nodal signaling in GBM therapy.

11.
Mol Neurobiol ; 52(1): 653-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25257696

ABSTRACT

Recent clinical studies have shown that sepsis survivors may develop long-term cognitive impairments. The cellular and molecular mechanisms involved in these events are not well understood. This study investigated synaptic deficits in sepsis and the involvement of glial cells in this process. Septic animals showed memory impairment and reduced numbers of hippocampal and cortical excitatory synapses, identified by synaptophysin/PSD-95 co-localization, 9 days after disease onset. The behavioral deficits and synaptophysin/PSD-95 co-localization were rescued to normal levels within 30 days post-sepsis. Septic mice presented activation of microglia and reactive astrogliosis, which are hallmarks of brain injury and could be involved in the associated synaptic deficits. We treated neuronal cultures with conditioned medium derived from cultured astrocytes (ACM) and microglia (MCM) that were either non-stimulated or stimulated with lipopolysaccharide (LPS) to investigate the molecular mechanisms underlying synaptic deficits in sepsis. ACM and MCM increased the number of synapses between cortical neurons in vitro, and these effects were antagonized by LPS stimulation. LPS-MCM reduced the number of synapses by 50%, but LPS-ACM increased the number of synapses by 500%. Analysis of the composition of these conditioned media revealed increased levels of IL-1ß in LPS-MCM. Furthermore, inhibition of IL-1ß signaling through the addition of a soluble IL-1ß receptor antagonist (IL-1 Ra) fully prevented the synaptic deficit induced by LPS-MCM. These results suggest that sepsis induces a transient synaptic deficit associated with memory impairments mediated by IL-1ß secreted by activated microglia.


Subject(s)
Cognition Disorders/etiology , Interleukin-1beta/metabolism , Microglia/pathology , Sepsis/complications , Synapses/pathology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cognition Disorders/pathology , Gliosis/etiology , Gliosis/pathology , Hippocampus/drug effects , Hippocampus/pathology , Lipopolysaccharides/pharmacology , Memory Disorders/etiology , Memory Disorders/pathology , Mice , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/pathology , Sepsis/pathology , Synapses/drug effects , Synapses/metabolism
12.
BMC Cancer ; 14: 923, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25482099

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common primary brain tumor and the most aggressive glial tumor. This tumor is highly heterogeneous, angiogenic, and insensitive to radio- and chemotherapy. Here we have investigated the progression of GBM produced by the injection of human GBM cells into the brain parenchyma of immunocompetent mice. METHODS: Xenotransplanted animals were submitted to magnetic resonance imaging (MRI) and histopathological analyses. RESULTS: Our data show that two weeks after injection, the produced tumor presents histopathological characteristics recommended by World Health Organization for the diagnosis of GBM in humans. The tumor was able to produce reactive gliosis in the adjacent parenchyma, angiogenesis, an intense recruitment of macrophage and microglial cells, and presence of necrosis regions. Besides, MRI showed that tumor mass had enhanced contrast, suggesting a blood-brain barrier disruption. CONCLUSIONS: This study demonstrated that the xenografted tumor in mouse brain parenchyma develops in a very similar manner to those found in patients affected by GBM and can be used to better understand the biology of GBM as well as testing potential therapies.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Disease Models, Animal , Glioblastoma/pathology , Tumor Microenvironment , Animals , Brain/blood supply , Brain Neoplasms/complications , Glioblastoma/complications , Glioblastoma/physiopathology , Gliosis/etiology , Humans , Immunocompetence , Macrophage Activation , Magnetic Resonance Imaging , Male , Mice , Microglia/physiology , Necrosis/etiology , Neovascularization, Pathologic/etiology , Transplantation, Heterologous
13.
Front Cell Neurosci ; 8: 362, 2014.
Article in English | MEDLINE | ID: mdl-25404894

ABSTRACT

The blood-brain barrier (BBB), constituted by an extensive network of endothelial cells (ECs) together with neurons and glial cells, including microglia, forms the neurovascular unit (NVU). The crosstalk between these cells guarantees a proper environment for brain function. In this context, changes in the endothelium-microglia interactions are associated with a variety of inflammation-related diseases in brain, where BBB permeability is compromised. Increasing evidences indicate that activated microglia modulate expression of tight junctions, which are essential for BBB integrity and function. On the other hand, the endothelium can regulate the state of microglial activation. Here, we review recent advances that provide insights into interactions between the microglia and the vascular system in brain diseases such as infectious/inflammatory diseases, epilepsy, ischemic stroke and neurodegenerative disorders.

14.
J Neuroimmunol ; 274(1-2): 71-7, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25042352

ABSTRACT

Factors released by glioma-associated microglia/macrophages (GAMs) play an important role in the growth and infiltration of tumors. We have previously demonstrated that the co-chaperone stress-inducible protein 1 (STI1) secreted by microglia promotes proliferation and migration of human glioblastoma (GBM) cell lines in vitro. In the present study, in order to investigate the role of STI1 in a physiological context, we used a glioma model to evaluate STI1 expression in vivo. Here, we demonstrate that STI1 expression in both the tumor and in the infiltrating GAMs and lymphocytes significantly increased with tumor progression. Interestingly, high expression of STI1 was observed in macrophages and lymphocytes that infiltrated brain tumors, whereas STI1 expression in the circulating blood monocytes and lymphocytes remained unchanged. Our results correlate, for the first time, the expression of STI1 and glioma progression, and suggest that STI1 expression in GAMs and infiltrating lymphocytes is modulated by the brain tumor microenvironment.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Heat-Shock Proteins/immunology , Macrophages/immunology , Microglia/immunology , Animals , Brain Neoplasms/metabolism , CX3C Chemokine Receptor 1 , Cell Line, Tumor , Disease Progression , Female , Flow Cytometry , Gene Expression/immunology , Glioma/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Chemokine/genetics , Tumor Microenvironment/immunology
15.
Clin Exp Metastasis ; 31(4): 461-74, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24488147

ABSTRACT

Tumor establishment, growth, and survival are supported by interactions with microenvironment components. Here, we investigated whether the interactions between prostate cancer cells and cortical astrocytes are associated to a potential role for astrocytes in tumor establishment. We demonstrate that astrocytes interact in vitro with prostatic cancers cells derived from different metastatic sites. Astrocytes and their secreted extracellular matrix, stimulate DU145 cell (a brain-derived prostate tumor cell line) proliferation while inhibiting cell death and modulating the expression of several genes related to prostate cancer progression, suggesting the activation of EMT process in these cells. In contrast, DU145 cells and their conditioned medium inhibited cell proliferation and induced cell death of astrocytes. On the other hand, the astrocytes were unable to significantly induce an increment of LNCaP cell (a lymph node-derived prostate tumor cell line) proliferative activity. In addition, LNCaP cells were also unable to induce cell death of astrocytes. Thus, we believe that DU145 cells, but not LNCaP cells, present an even more aggressive behavior when interacting with astrocytes. These results provide an important contribution to the elucidation of the cellular mechanisms involved in the brain microenvironment colonization.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/secondary , Cell Communication , Cell Movement , Prostatic Neoplasms/pathology , Apoptosis , Astrocytes/metabolism , Brain Neoplasms/genetics , Cell Proliferation , Gene Expression Profiling , Humans , Male , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Microenvironment
16.
FEBS Lett ; 587(2): 238-44, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23238082

ABSTRACT

Prion protein (PrP(C)) has neuroprotective functions and herein we demonstrate that astrocytes from PrP(C)-over-expressing mice are more resistant to induced cell death than wild-type astrocytes. The Stress-Inducible-Protein 1 (STI1), a PrP(C) ligand, prevents cell death in both wild-type and PrP(C)-over-expressing astrocytes through the activation of protein-kinase-A. Cultured embryonic astrocytes and brain extracts from PrP(C)-over-expressing mice show higher glial fibrillary acidic protein expression and reduced vimentin and nestin levels when compared to wild-type astrocytes, suggesting faster astrocyte maturation in the former mice. Our data indicate that PrP(C) levels modulate astrocyte development, and that PrP(C)-STI1 interaction contributes to protect against astrocyte death.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , PrPC Proteins/metabolism , Animals , Cell Death/genetics , Cell Death/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Glial Fibrillary Acidic Protein , Heat-Shock Proteins/metabolism , Intermediate Filament Proteins/metabolism , MAP Kinase Signaling System , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nestin , PrPC Proteins/genetics , Up-Regulation , Vimentin/metabolism
17.
Life Sci ; 89(15-16): 532-9, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21641917

ABSTRACT

Glioblastomas (GBMs) are considered to be one of the deadliest human cancers, characterized by a high proliferative rate, aggressive invasiveness and insensitivity to radio- and chemotherapy, as well as a short patient survival period. Moreover, GBMs are among the most vascularized and invasive cancers in humans. Angiogenesis in GBMs is correlated with the grade of malignancy and is inversely correlated with patient survival. One of the first steps in tumor invasions is migration. GBM cells have the ability to infiltrate and disrupt physical barriers such as basement membranes, extracellular matrix and cell junctions. The invasion process includes the overexpression of several members of a super-family of zinc-based proteinases, the Metzincin, in particular a sub-group, metalloproteinases. Another interesting aspect is that, inside the GBM tissue, there are up to 30% of microglia or macrophages. However, little is known about the immune performance and interactions of the microglia with GBMs. These singular properties of GBMs will be described here. A sub-population of cells with stem-like properties may be the source of tumors since, apparently, GBM stem cells (GSCs) are highly resistant to current cancer treatments. These cancer therapies, while killing the majority of tumor cells, ultimately fail in GBM treatment because they do not eliminate GSCs, which survive to regenerate new tumors. Finally, GBM patient prognostic has shown little improvement in decades. In this context, we will discuss how the membrane-acting toxins called cytolysins can be a potential new tool for GBM treatment.


Subject(s)
Glioblastoma/pathology , Nervous System Neoplasms/pathology , Animals , Glioblastoma/blood supply , Humans , Metalloproteases/physiology , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/physiology , Neovascularization, Pathologic/pathology , Nervous System Neoplasms/blood supply , Pore Forming Cytotoxic Proteins/metabolism
18.
Cell Tissue Res ; 342(2): 179-89, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20878337

ABSTRACT

In a previous study, we analyzed and described the features of the degeneration of the protocerebral tract (PCT) of the crustacean Ucides cordatus, after the extirpation of the eyestalk. In that study, among axons with axoplasmic degeneration, cells with granules resembling blood cells (hemocytes) were seen. Therefore, in the present study, we characterized the circulating hemocytes and compared them with the cells recruited to a lesion, which was produced as in the former study. Using histochemistry, immunohistochemistry, and electron microscopy (transmission and scanning), we confirmed that circulating and recruited cells display a similar morphology. Therefore, in the crab, hemocytes were attracted to the lesion site in the acute stage of degeneration, appearing near local glial cells that showed signs of being responsive. Some of the attracted hemocytes displayed a morphology that was considered to be possibly activated blood cells. Also, the cells that migrated to the injured PCT displayed features, such as the presence of hydrolytic enzymes and an ability to phagocytize neural debris, similar to those of vertebrates. In summary, our results indicate that hemocytes were not only phagocytizing neural debris together with glial cells but also that they may be concerned with creating a favorable environment for regenerating events.


Subject(s)
Brachyura , Central Nervous System/pathology , Hemocytes/pathology , Optic Lobe, Nonmammalian/pathology , Wallerian Degeneration/pathology , Animals , Axons/metabolism , Axons/pathology , Axons/ultrastructure , Cell Movement , Central Nervous System/injuries , Hemocytes/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nerve Regeneration/physiology , Neuroglia/pathology , Neuroglia/ultrastructure , Optic Lobe, Nonmammalian/metabolism , Optic Lobe, Nonmammalian/ultrastructure , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL
...