Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Neuroinflammation ; 21(1): 161, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38915059

RÉSUMÉ

BACKGROUND: Pediatric acute transverse myelitis (ATM) accounts for 20-30% of children presenting with a first acquired demyelinating syndrome (ADS) and may be the first clinical presentation of a relapsing ADS such as multiple sclerosis (MS). B cells have been strongly implicated in the pathogenesis of adult MS. However, little is known about B cells in pediatric MS, and even less so in pediatric ATM. Our lab previously showed that plasmablasts (PB), the earliest B cell subtype producing antibody, are expanded in adult ATM, and that these PBs produce self-reactive antibodies that target neurons. The goal of this study was to examine PB frequency and phenotype, immunoglobulin selection, and B cell receptor reactivity in pediatric patients presenting with ATM to gain insight to B cell involvement in disease. METHODS: We compared the PB frequency and phenotype of 5 pediatric ATM patients and 10 pediatric healthy controls (HC) and compared them to previously reported adult ATM patients using cytometric data. We purified bulk IgG from the plasma samples and cloned 20 recombinant human antibodies (rhAbs) from individual PBs isolated from the blood. Plasma-derived IgG and rhAb autoreactivity was measured by mean fluorescence intensity (MFI) in neurons and astrocytes of murine brain or spinal cord and primary human astrocytes. We determined the potential impact of these rhAbs on astrocyte health by measuring stress and apoptotic response. RESULTS: We found that pediatric ATM patients had a reduced frequency of peripheral blood PB. Serum IgG autoreactivity to neurons in EAE spinal cord was similar in the pediatric ATM patients and HC. However, serum IgG autoreactivity to astrocytes in EAE spinal cord was reduced in pediatric ATM patients compared to pediatric HC. Astrocyte-binding strength of rhAbs cloned from PBs was dependent on somatic hypermutation accumulation in the pediatric ATM cohort, but not HC. A similar observation in predilection for astrocyte binding over neuron binding of individual antibodies cloned from PBs was made in EAE brain tissue. Finally, exposure of human primary astrocytes to these astrocyte-binding antibodies increased astrocytic stress but did not lead to apoptosis. CONCLUSIONS: Discordance in humoral immune responses to astrocytes may distinguish pediatric ATM from HC.


Sujet(s)
Astrocytes , Myélite transverse , Humains , Myélite transverse/immunologie , Animaux , Femelle , Astrocytes/métabolisme , Astrocytes/immunologie , Enfant , Souris , Mâle , Adolescent , Plasmocytes/immunologie , Plasmocytes/métabolisme , Autoanticorps/immunologie , Autoanticorps/sang , Souris de lignée C57BL , Cellules cultivées , Enfant d'âge préscolaire , Immunoglobuline G/immunologie , Immunoglobuline G/sang , Moelle spinale/métabolisme , Moelle spinale/immunologie , Moelle spinale/anatomopathologie
2.
Bioinformatics ; 39(9)2023 09 02.
Article de Anglais | MEDLINE | ID: mdl-37624931

RÉSUMÉ

MOTIVATION: As an important player in transcriptome regulation, microRNAs may effectively diffuse somatic mutation impacts to broad cellular processes and ultimately manifest disease and dictate prognosis. Previous studies that tried to correlate mutation with gene expression dysregulation neglected to adjust for the disparate multitudes of false positives associated with unequal sample sizes and uneven class balancing scenarios. RESULTS: To properly address this issue, we developed a statistical framework to rigorously assess the extent of mutation impact on microRNAs in relation to a permutation-based null distribution of a matching sample structure. Carrying out the framework in a pan-cancer study, we ascertained 9008 protein-coding genes with statistically significant mutation impacts on miRNAs. Of these, the collective miRNA expression for 83 genes showed significant prognostic power in nine cancer types. For example, in lower-grade glioma, 10 genes' mutations broadly impacted miRNAs, all of which showed prognostic value with the corresponding miRNA expression. Our framework was further validated with functional analysis and augmented with rich features including the ability to analyze miRNA isoforms; aggregative prognostic analysis; advanced annotations such as mutation type, regulator alteration, somatic motif, and disease association; and instructive visualization such as mutation OncoPrint, Ideogram, and interactive mRNA-miRNA network. AVAILABILITY AND IMPLEMENTATION: The data underlying this article are available in MutMix, at http://innovebioinfo.com/Database/TmiEx/MutMix.php.


Sujet(s)
Gliome , microARN , Humains , Diffusion , microARN/génétique , Mutation , ARN messager
3.
Mol Cancer Res ; 21(8): 755-767, 2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-37255362

RÉSUMÉ

The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and recurrence, and improve the molecular classification of GBM.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Adulte , Humains , Glioblastome/anatomopathologie , Fluorescence , Tumeurs du cerveau/anatomopathologie , Microenvironnement tumoral/génétique
4.
Mol Cancer Res ; 21(8): 755-767, 2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-37159022

RÉSUMÉ

The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and the recurrence, and improve the molecular classification of GBM.

5.
Curr Med Chem ; 28(4): 687-711, 2021.
Article de Anglais | MEDLINE | ID: mdl-31830881

RÉSUMÉ

Melanoma is the most life-threatening and aggressive class of skin malignancies. The incidence of melanoma has steadily increased. Metastatic melanoma is greatly resistant to standard antimelanoma treatments such as chemotherapy, and the 5-year survival rate of cases with melanoma who have a metastatic form of the disease is less than 10%. The contributing role of apoptosis, angiogenesis and autophagy in the pathophysiology of melanoma has been previously demonstrated. Thus, it is extremely urgent to search for complementary therapeutic approaches that could enhance the quality of life of subjects and reduce treatment resistance and adverse effects. Resveratrol, known as a polyphenol component present in grapes and some plants, has anti-cancer properties due to its function as an apoptosis inducer in tumor cells, and anti-angiogenic agent to prevent metastasis. However, more clinical trials should be conducted to prove resveratrol efficacy. Herein, for the first time, we summarize the current knowledge of anti-cancerous activities of resveratrol in melanoma.


Sujet(s)
Mélanome , Tumeurs cutanées , Stilbènes , Apoptose , Humains , Mélanome/traitement médicamenteux , Qualité de vie , Resvératrol/pharmacologie , Resvératrol/usage thérapeutique , Tumeurs cutanées/traitement médicamenteux , Stilbènes/pharmacologie , Stilbènes/usage thérapeutique
6.
Cell Rep ; 30(8): 2489-2500.e5, 2020 02 25.
Article de Anglais | MEDLINE | ID: mdl-32101730

RÉSUMÉ

Emerging evidence suggests that crosstalk between glioma cells and the brain microenvironment may influence brain tumor growth. To date, known reciprocal interactions among these cells have been limited to the release of paracrine factors. Combining a genetic strategy with longitudinal live imaging, we find that individual gliomas communicate with distinct sets of non-glioma cells, including glial cells, neurons, and vascular cells. Transfer of genetic material is achieved mainly through extracellular vesicles (EVs), although cell fusion also plays a minor role. We further demonstrate that EV-mediated communication leads to the increase of synaptic activity in neurons. Blocking EV release causes a reduction of glioma growth in vivo. Our findings indicate that EV-mediated interaction between glioma cells and non-glioma brain cells alters the tumor microenvironment and contributes to glioma development.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Encéphale/anatomopathologie , Communication cellulaire , Vésicules extracellulaires/métabolisme , Gliome/anatomopathologie , Animaux , Astrocytes/anatomopathologie , Encéphale/physiopathologie , Tumeurs du cerveau/physiopathologie , Fusion cellulaire , Lignée cellulaire tumorale , ADN tumoral/génétique , Phénomènes électrophysiologiques , Vésicules extracellulaires/ultrastructure , Gliome/physiopathologie , Humains , Souris de lignée C57BL , Souris nude , Neurones/anatomopathologie , Imagerie accélérée
7.
J Neuropathol Exp Neurol ; 78(12): 1081-1088, 2019 12 01.
Article de Anglais | MEDLINE | ID: mdl-31589317

RÉSUMÉ

Clear cell, microcytic, and angiomatous meningiomas are 3 vasculature-rich variants with overlapping morphological features but different prognostic and treatment implications. Distinction between them is not always straightforward. We compared the expression patterns of the hypoxia marker carbonic anhydrase IX (CA-IX) in meningiomas with predominant clear cell (n = 15), microcystic (n = 9), or angiomatous (n = 11) morphologies, as well as 117 cases of other World Health Organization recognized histological meningioma variants. Immunostaining for SMARCE1 protein, whose loss-of-function has been associated with clear cell meningiomas, was performed on all clear cell meningiomas, and selected variants of meningiomas as controls. All clear cell meningiomas showed absence of CA-IX expression and loss of nuclear SMARCE1 expression. All microcystic and angiomatous meningiomas showed diffuse CA-IX immunoreactivity and retained nuclear SMARCE1 expression. In other meningioma variants, CA-IX was expressed in a hypoxia-restricted pattern and was highly associated with atypical features such as necrosis, small cell change, and focal clear cell change. In conclusion, CA-IX may serve as a useful diagnostic marker in differentiating clear cell, microcystic, and angiomatous meningiomas.


Sujet(s)
Antigènes néoplasiques/métabolisme , Carbonic anhydrase IX/métabolisme , Tumeurs des méninges/enzymologie , Méningiome/enzymologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/métabolisme , Encéphale/anatomopathologie , Protéines chromosomiques nonhistones/métabolisme , Protéines de liaison à l'ADN/métabolisme , Femelle , Humains , Mâle , Tumeurs des méninges/diagnostic , Tumeurs des méninges/anatomopathologie , Méningiome/diagnostic , Méningiome/anatomopathologie , Adulte d'âge moyen , Survie sans progression
8.
Neuro Oncol ; 21(6): 719-729, 2019 06 10.
Article de Anglais | MEDLINE | ID: mdl-30590711

RÉSUMÉ

BACKGROUND: Expression of neuron-glial antigen 2 (NG2) identifies an aggressive malignant phenotype in glioblastoma (GBM). Mouse models have implicated NG2 in the genesis, evolution, and maintenance of glial cancers and have highlighted potential interactions between NG2 and epidermal growth factor receptor (EGFR). However, it is unknown whether the lineage relationship of NG2+ and NG2- cells follows a hierarchical or stochastic mode of growth. Furthermore, the interaction between NG2 and EGFR signaling in human GBM is also unclear. METHODS: Single GBM NG2+ and NG2- cells were studied longitudinally to assess lineage relationships. Short hairpin RNA knockdown of NG2 was used to assess the mechanistic role of NG2 in human GBM cells. NG2+ and NG2- cells and NG2 knockdown (NG2-KD) and wild type (NG2-WT) cells were analyzed for differential effects on EGFR signaling. RESULTS: Expression of NG2 endows an aggressive phenotype both at single cell and population levels. Progeny derived from single GBM NG2- or GBM NG2+ cells consistently establish phenotypic equilibrium, indicating the absence of a cellular hierarchy. NG2 knockdown reduces proliferation, and mice grafted with NG2-KD survive longer than controls. Finally, NG2 promotes EGFR signaling and is associated with EGFR expression. CONCLUSIONS: These data support a dynamic evolution in which a bidirectional relationship exists between GBM NG2+ and GBM NG2- cells. Such findings have implications for understanding phenotypic heterogeneity, the emergence of resistant disease, and developing novel therapeutics.


Sujet(s)
Antigènes/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du cerveau/anatomopathologie , Régulation de l'expression des gènes tumoraux , Glioblastome/anatomopathologie , Protéoglycanes/métabolisme , Animaux , Antigènes/génétique , Apoptose , Marqueurs biologiques tumoraux/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Carcinogenèse , Prolifération cellulaire , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Glioblastome/génétique , Glioblastome/métabolisme , Humains , Souris , Protéoglycanes/génétique , Transduction du signal , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
11.
CNS Oncol ; 4(6): 399-410, 2015.
Article de Anglais | MEDLINE | ID: mdl-26497327

RÉSUMÉ

Glioblastoma demonstrates imaging features of intratumor heterogeneity that result from underlying heterogeneous biological properties. This stems from variations in cellular behavior that result from genetic mutations that either drive, or are driven by, heterogeneous microenvironment conditions. Among all imaging methods available, only T1-weighted contrast-enhancing and T2-weighted fluid-attenuated inversion recovery are used in standard clinical glioblastoma assessment and monitoring. Advanced imaging modalities are still considered emerging techniques as appropriate end points and robust methodologies are missing from clinical trials. Discovering how these images specifically relate to the underlying tumor biology may aid in improving quality of clinical trials and understanding the factors involved in regional responses to treatment, including variable drug uptake and effect of radiotherapy. Upon validation and standardization of emerging MR techniques, providing information based on the underlying tumor biology, these images may allow for clinical decision-making that is tailored to an individual's response to treatment.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Glioblastome/anatomopathologie , Imagerie multimodale/méthodes , Animaux , Tumeurs du cerveau/thérapie , Glioblastome/thérapie , Humains
12.
Future Oncol ; 11(20): 2851, 2015.
Article de Anglais | MEDLINE | ID: mdl-26358184
13.
Future Oncol ; 11(12): 1703-6, 2015.
Article de Anglais | MEDLINE | ID: mdl-26075436

RÉSUMÉ

"Serial tumor sampling, single-cell genomics and quantitative imaging are all available technologies, but their integration into current pathways of care will require a paradigm shift in the clinical management of patients with glioblastoma."


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Glioblastome/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/thérapie , Évolution de la maladie , Glioblastome/génétique , Glioblastome/thérapie , Humains , Thérapie moléculaire ciblée , Mutation , Transduction du signal
14.
Stem Cells Transl Med ; 4(7): 822-31, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-26019225

RÉSUMÉ

UNLABELLED: : Recent research has focused on the hypothesis that the growth and regeneration of glioblastoma (GB) is sustained by a subpopulation of self-renewing stem-like cells. This has led to the prediction that molecular markers for cancer stem cells in GB may provide a treatment target. One candidate marker is CD15: we wanted to determine if CD15 represented a credible stem cell marker in GB. We first demonstrated that CD15-positive (CD15+) cells were less proliferative than their CD15-negative (CD15-) counterparts in 10 patient GB tumors. Next we compared the proliferative activity of CD15+ and CD15- cells in vitro using tumor-initiating primary GB cell lines (TICs) and found no difference in proliferative behavior. Furthermore, TICs sorted for CD15+ and CD15- were not significantly different cytogenetically or in terms of gene expression profile. Sorted single CD15+ and CD15- cells were equally capable of reconstituting a heterogeneous population containing both CD15+ and CD15- cells over time, and both CD15+ and CD15- cells were able to generate tumors in vivo. No difference was found in the phenotypic or genomic behavior of CD15+ cells compared with CD15- cells from the same patient. Moreover, we found that in vitro, cells were able to interconvert between the CD15+ and CD15- states. Our data challenge the utility of CD15 as a cancer stem cell marker. SIGNIFICANCE: The data from this study contribute to the ongoing debate about the role of cancer stem cells in gliomagenesis. Results showed that CD15, a marker previously thought to be a cancer stem-like marker in glioblastoma, could not isolate a phenotypically or genetically distinct population. Moreover, isolated CD15-positive and -negative cells were able to generate mixed populations of glioblastoma cells in vitro.

15.
Stem Cell Reports ; 4(1): 7-15, 2015 Jan 13.
Article de Anglais | MEDLINE | ID: mdl-25533637

RÉSUMÉ

Glioblastoma (GBM) is a lethal malignancy whose clinical intransigence has been linked to extensive intraclonal genetic and phenotypic diversity and the common emergence of therapeutic resistance. This interpretation embodies the implicit assumption that cancer stem cells or tumor-propagating cells are themselves genetically and functionally diverse. To test this, we screened primary GBM tumors by SNP array to identify copy number alterations (a minimum of three) that could be visualized in single cells by multicolor fluorescence in situ hybridization. Interrogation of neurosphere-derived cells (from four patients) and cells derived from secondary transplants of these same cells in NOD-SCID mice allowed us to infer the clonal and phylogenetic architectures. Whole-exome sequencing and single-cell genetic analysis in one case revealed a more complex clonal structure. This proof-of-principle experiment revealed that subclones in each GBM had variable regenerative or stem cell activity, and highlighted genetic alterations associated with more competitive propagating activity in vivo.


Sujet(s)
Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Variation génétique , Glioblastome/génétique , Glioblastome/métabolisme , Phénotype , Animaux , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Variations de nombre de copies de segment d'ADN , Évolution de la maladie , Étude d'association pangénomique , Génomique , Glioblastome/anatomopathologie , Hétérogreffes , Séquençage nucléotidique à haut débit , Humains , Hybridation fluorescente in situ , Souris , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Polymorphisme de nucléotide simple , Analyse sur cellule unique
16.
Proc Natl Acad Sci U S A ; 110(10): 4009-14, 2013 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-23412337

RÉSUMÉ

Glioblastoma (GB) is the most common and aggressive primary brain malignancy, with poor prognosis and a lack of effective therapeutic options. Accumulating evidence suggests that intratumor heterogeneity likely is the key to understanding treatment failure. However, the extent of intratumor heterogeneity as a result of tumor evolution is still poorly understood. To address this, we developed a unique surgical multisampling scheme to collect spatially distinct tumor fragments from 11 GB patients. We present an integrated genomic analysis that uncovers extensive intratumor heterogeneity, with most patients displaying different GB subtypes within the same tumor. Moreover, we reconstructed the phylogeny of the fragments for each patient, identifying copy number alterations in EGFR and CDKN2A/B/p14ARF as early events, and aberrations in PDGFRA and PTEN as later events during cancer progression. We also characterized the clonal organization of each tumor fragment at the single-molecule level, detecting multiple coexisting cell lineages. Our results reveal the genome-wide architecture of intratumor variability in GB across multiple spatial scales and patient-specific patterns of cancer evolution, with consequences for treatment design.


Sujet(s)
Tumeurs du cerveau/génétique , Glioblastome/génétique , Séquence nucléotidique , Aberrations des chromosomes , Variations de nombre de copies de segment d'ADN , ADN tumoral/génétique , Évolution de la maladie , Évolution moléculaire , Gènes erbB-1 , Gènes p16 , Humains , Phylogenèse , Transcriptome
17.
Acta Neurochir (Wien) ; 154(6): 951-69; discussion 969, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22527576

RÉSUMÉ

The neurosurgical management of patients with intrinsic glial cancers is one of the most rapidly evolving areas of practice. This has been fuelled by advances in surgical technique not only in cytoreduction but also in drug delivery. Further innovation will depend on a deeper understanding of the biology of the disease and an appreciation of the limitations of current knowledge. Here we review the controversial topic of cancer stem cells applied to glioma to provide neurosurgeons with a working overview. It is now recognised that the adult human brain contains regionally specified cell populations capable of self-renewal that may contribute to tumour growth and maintenance following accumulated mutational change. Tumour cells adapted to maintain growth demonstrate some stem-like characteristics and as such constitute a legitimate therapeutic target. Cellular reprogramming technologies raise the potential of developing stem cells as novel surgical tools to target disease and possibly ameliorate some of the consequences of treatment. Achieving these goals remains a significant challenge to neurosurgical oncologists, not least in challenging how we think about treating brain cancer. This review will briefly examine our understanding of adult stem cells within the brain, the evidence that they contribute to the development of brain tumours as tumour-initiating cells, and the potential implications for therapy. It will also look at the role stem cells may play in the future management of glioma.


Sujet(s)
Tumeurs du cerveau/étiologie , Tumeurs du cerveau/anatomopathologie , Gliome/thérapie , Transplantation de cellules souches/tendances , Animaux , Tumeurs du cerveau/thérapie , Différenciation cellulaire/physiologie , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Gliome/étiologie , Gliome/physiopathologie , Humains , Cellules souches tumorales/anatomopathologie , Transplantation de cellules souches/méthodes
18.
J Mol Med (Berl) ; 87(11): 1087-95, 2009 Nov.
Article de Anglais | MEDLINE | ID: mdl-19784875

RÉSUMÉ

Cancers comprise heterogeneous cells, ranging from highly proliferative immature precursors to more differentiated cell lineages. In the last decade, several groups have demonstrated the existence of cancer stem cells in both nonsolid solid tumors, including some of the brain: glioblastoma multiforme (GBM), medulloblastoma, and ependymoma. These cells, like their normal counterpart in homologous tissues, are multipotent, undifferentiated, self-sustaining, yet transformed cells. In particular, glioblastoma-stem like cells (GBSCs) self-renew under clonal conditions and differentiate into neuron- and glia-like cells, with aberrant, mixed neuronal/astroglial phenotypes. Remarkably, upon subcutaneous and intracerebral transplantation in immunosuppressed mice, GBSCs are able to form secondary tumors that closely resemble the human pathology, a property retained also throughout serial transplantation. The search is up for the identification of the markers and the molecular mechanisms that underpin the tumorigenic potential of these cells. This is critical if we aim at defining new therapeutic approaches for the treatment of malignant brain tumors. Lately, it has been shown that some key regulatory system that plays pivotal roles in neural stem cell physiology can also regulate the tumorigenic ability of cancer stem cells in GBMs. This suggests that the study of cancer stem cells in brain tumors might help to identify new and more specific therapeutic molecular effectors, with the cancer stem cells themselves representing one of the main targets, in fact the Holy Grail, in cancer cell therapy. This review includes a summary review on brain cancer cells and their usefulness as emerging targets in cancer cell therapy.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Encéphale/cytologie , Encéphale/anatomopathologie , Cellules souches tumorales/anatomopathologie , Animaux , Tumeurs du cerveau/thérapie , Thérapie cellulaire et tissulaire , Humains
19.
Expert Opin Biol Ther ; 7(8): 1129-35, 2007 Aug.
Article de Anglais | MEDLINE | ID: mdl-17696813

RÉSUMÉ

Cancers are composed of heterogeneous cell populations, including highly proliferative immature precursors and differentiated cells, which may belong to different lineages. Recent advances in stem cell research have demonstrated the existence of tumour-initiating, cancer stem cells (CSCs) in non-solid and solid tumours. These cells are defined as CSCs because they show functional properties that resemble those of their normal counterpart to a significant extent. This concept applies to CSCs from brain tumours and, particularly, to glioblastoma stem-like cells, which self-renew under clonal conditions and differentiate into neuron- and glia-like cells, and into aberrant cells, with mixed neuronal/astroglia phenotypes. Notably, across serial transplantation into immunodeficient mice, glioblastoma stem-like cells are able to form secondary tumours which are a phenocopy of the human disease. A significant effort is underway to identify both CSC-specific markers and the molecular mechanism that underpin the tumorigenic potential of these cells, for this will have a critical impact on the understanding of the origin of malignant brain tumour and the discovery of new and more specific therapeutic approaches. Lately, the authors have shown that some of the bone morphogenetic proteins can reduce the tumorigenic ability of CSCs in GBMs. This suggests that mechanisms regulating the physiology of normal brain stem cells may be still in place in their cancerous siblings and that this may lead to the development of cures that selectively target the population CSCs found in the patients' tumour mass.


Sujet(s)
Tumeurs du cerveau/thérapie , Cellules souches tumorales/transplantation , Animaux , Tumeurs du cerveau/anatomopathologie , Transformation cellulaire néoplasique/anatomopathologie , Humains , Transplantation tumorale/méthodes , Cellules souches tumorales/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...