Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 174, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39014482

RESUMEN

BACKGROUND: Specific microglia responses are thought to contribute to the development and progression of neurodegenerative diseases, including Parkinson's disease (PD). However, the phenotypic acquisition of microglial cells and their role during the underlying neuroinflammatory processes remain largely elusive. Here, according to the multiple-hit hypothesis, which stipulates that PD etiology is determined by a combination of genetics and various environmental risk factors, we investigate microglial transcriptional programs and morphological adaptations under PARK7/DJ-1 deficiency, a genetic cause of PD, during lipopolysaccharide (LPS)-induced inflammation. METHODS: Using a combination of single-cell RNA-sequencing, bulk RNA-sequencing, multicolor flow cytometry and immunofluorescence analyses, we comprehensively compared microglial cell phenotypic characteristics in PARK7/DJ-1 knock-out (KO) with wildtype littermate mice following 6- or 24-h intraperitoneal injection with LPS. For translational perspectives, we conducted corresponding analyses in human PARK7/DJ-1 mutant induced pluripotent stem cell (iPSC)-derived microglia and murine bone marrow-derived macrophages (BMDMs). RESULTS: By excluding the contribution of other immune brain resident and peripheral cells, we show that microglia acutely isolated from PARK7/DJ-1 KO mice display a distinct phenotype, specially related to type II interferon and DNA damage response signaling, when compared with wildtype microglia, in response to LPS. We also detected discrete signatures in human PARK7/DJ-1 mutant iPSC-derived microglia and BMDMs from PARK7/DJ-1 KO mice. These specific transcriptional signatures were reflected at the morphological level, with microglia in LPS-treated PARK7/DJ-1 KO mice showing a less amoeboid cell shape compared to wildtype mice, both at 6 and 24 h after acute inflammation, as also observed in BMDMs. CONCLUSIONS: Taken together, our results show that, under inflammatory conditions, PARK7/DJ-1 deficiency skews microglia towards a distinct phenotype characterized by downregulation of genes involved in type II interferon signaling and a less prominent amoeboid morphology compared to wildtype microglia. These findings suggest that the underlying oxidative stress associated with the lack of PARK7/DJ-1 affects microglia neuroinflammatory responses, which may play a causative role in PD onset and progression.


Asunto(s)
Inflamación , Lipopolisacáridos , Ratones Noqueados , Microglía , Proteína Desglicasa DJ-1 , Animales , Proteína Desglicasa DJ-1/deficiencia , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo , Microglía/metabolismo , Microglía/patología , Microglía/efectos de los fármacos , Ratones , Lipopolisacáridos/toxicidad , Lipopolisacáridos/farmacología , Inflamación/patología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/genética , Humanos , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/genética
2.
Genome Med ; 16(1): 51, 2024 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-38566128

RESUMEN

BACKGROUND: A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. METHODS: Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry, and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. RESULTS: We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood-brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. CONCLUSIONS: Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Ratones , Animales , Humanos , Glioblastoma/genética , Glioblastoma/metabolismo , Microglía/metabolismo , Ecosistema , Xenoinjertos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Fenotipo , Modelos Animales de Enfermedad , Células Dendríticas/metabolismo , Microambiente Tumoral/genética
3.
Cancer Gene Ther ; 30(10): 1330-1345, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37420093

RESUMEN

Therapy Induced Senescence (TIS) leads to sustained growth arrest of cancer cells. The associated cytostasis has been shown to be reversible and cells escaping senescence further enhance the aggressiveness of cancers. Chemicals specifically targeting senescent cells, so-called senolytics, constitute a promising avenue for improved cancer treatment in combination with targeted therapies. Understanding how cancer cells evade senescence is needed to optimise the clinical benefits of this therapeutic approach. Here we characterised the response of three different NRAS mutant melanoma cell lines to a combination of CDK4/6 and MEK inhibitors over 33 days. Transcriptomic data show that all cell lines trigger a senescence programme coupled with strong induction of interferons. Kinome profiling revealed the activation of Receptor Tyrosine Kinases (RTKs) and enriched downstream signaling of neurotrophin, ErbB and insulin pathways. Characterisation of the miRNA interactome associates miR-211-5p with resistant phenotypes. Finally, iCell-based integration of bulk and single-cell RNA-seq data identifies biological processes perturbed during senescence and predicts 90 new genes involved in its escape. Overall, our data associate insulin signaling with persistence of a senescent phenotype and suggest a new role for interferon gamma in senescence escape through the induction of EMT and the activation of ERK5 signaling.


Asunto(s)
Insulinas , Melanoma , Humanos , Multiómica , Línea Celular Tumoral , Melanoma/tratamiento farmacológico , Melanoma/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Insulinas/uso terapéutico , Senescencia Celular/genética , Proteínas de la Membrana/genética , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/uso terapéutico
4.
EMBO Mol Med ; 15(8): e17241, 2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37278126

RESUMEN

In early life, the intestinal mucosa and immune system undergo a critical developmental process to contain the expanding gut microbiome while promoting tolerance toward commensals, yet the influence of maternal diet and microbial composition on offspring immune maturation remains poorly understood. We colonized germ-free mice with a consortium of 14 strains, fed them a standard fiber-rich chow or a fiber-free diet, and then longitudinally assessed offspring development during the weaning period. Unlike pups born to dams fed the fiber-rich diet, pups of fiber-deprived dams demonstrated delayed colonization with Akkermansia muciniphila, a mucin-foraging bacterium that can also use milk oligosaccharides. The pups of fiber-deprived dams exhibited an enrichment of colonic transcripts corresponding to defense response pathways and a peak in Il22 expression at weaning. Removal of A. muciniphila from the community, but maintenance on the fiber-rich diet, was associated with reduced proportions of RORγt-positive innate and adaptive immune cell subsets. Our results highlight the potent influence of maternal dietary fiber intake and discrete changes in microbial composition on the postnatal microbiome assemblage and early immune development.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Ratones , Animales , Dieta , Mucosa Intestinal , Colon
5.
bioRxiv ; 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36945572

RESUMEN

Background: A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. Methods: Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA-sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. Results: We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood-brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. Conclusions: Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.

6.
Allergy ; 78(3): 682-696, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36210648

RESUMEN

BACKGROUND: Numerous patient-based studies have highlighted the protective role of immunoglobulin E-mediated allergic diseases on glioblastoma (GBM) susceptibility and prognosis. However, the mechanisms behind this observation remain elusive. Our objective was to establish a preclinical model able to recapitulate this phenomenon and investigate the role of immunity underlying such protection. METHODS: An immunocompetent mouse model of allergic airway inflammation (AAI) was initiated before intracranial implantation of mouse GBM cells (GL261). RAG1-KO mice served to assess tumor growth in a model deficient for adaptive immunity. Tumor development was monitored by MRI. Microglia were isolated for functional analyses and RNA-sequencing. Peripheral as well as tumor-associated immune cells were characterized by flow cytometry. The impact of allergy-related microglial genes on patient survival was analyzed by Cox regression using publicly available datasets. RESULTS: We found that allergy establishment in mice delayed tumor engraftment in the brain and reduced tumor growth resulting in increased mouse survival. AAI induced a transcriptional reprogramming of microglia towards a pro-inflammatory-like state, uncovering a microglia gene signature, which correlated with limited local immunosuppression in glioma patients. AAI increased effector memory T-cells in the circulation as well as tumor-infiltrating CD4+ T-cells. The survival benefit conferred by AAI was lost in mice devoid of adaptive immunity. CONCLUSION: Our results demonstrate that AAI limits both tumor take and progression in mice, providing a preclinical model to study the impact of allergy on GBM susceptibility and prognosis, respectively. We identify a potentiation of local and adaptive systemic immunity, suggesting a reciprocal crosstalk that orchestrates allergy-induced immune protection against GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Hipersensibilidad , Ratones , Animales , Glioblastoma/genética , Glioblastoma/patología , Neoplasias Encefálicas/patología , Glioma/genética , Glioma/patología , Microglía/patología , Hipersensibilidad/patología , Ratones Endogámicos C57BL
7.
Ann Endocrinol (Paris) ; 83(6): 423-430, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36306894

RESUMEN

The SFE-AFCE-SFMN 2022 consensus deals with the management of thyroid nodules, a condition that is a frequent reason for consultation in endocrinology. In more than 90% of cases, patients are euthyroid, with benign non-progressive nodules that do not warrant specific treatment. The clinician's objective is to detect malignant thyroid nodules at risk of recurrence and death, toxic nodules responsible for hyperthyroidism or compressive nodules warranting treatment. The diagnosis and treatment of thyroid nodules requires close collaboration between endocrinologists, nuclear medicine physicians and surgeons, but also involves other specialists. Therefore, this consensus statement was established jointly by 3 societies: the French Society of Endocrinology (SFE), French Association of Endocrine Surgery (AFCE) and French Society of Nuclear Medicine (SFMN); the various working groups included experts from other specialties (pathologists, radiologists, pediatricians, biologists, etc.). This section deals with thermal ablation, which may constitute an alternative to thyroid surgery in selected patients.


Asunto(s)
Endocrinología , Hipertiroidismo , Medicina Nuclear , Nódulo Tiroideo , Humanos , Nódulo Tiroideo/diagnóstico , Nódulo Tiroideo/cirugía , Resultado del Tratamiento
8.
Can Vet J ; 63(10): 1027-1031, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36185788

RESUMEN

A 4-month-old male beagle dog was presented for a 15-day history of firm cutaneous nodules. Histopathological examination of skin biopsies revealed calcinosis cutis. However, re-evaluation 40 d later confirmed spontaneous resolution of the lesions without specific treatment. Two weeks before development of the skin lesions, this dog had been hospitalized and treated for acute renal and hepatic disease attributed to leptospirosis, with both PCR and serology positive for Leptospira australis. Calcinosis cutis secondary to a systemic disease (leptospirosis, blastomycosis) has been rarely reported. In this case, the suspected pathogenesis included organic stress (cortisol hypersecretion) and abnormal calcium/phosphorus metabolism during acute renal failure. To our knowledge, this is the third published case of cutis calcinosis associated with leptospirosis in dogs. Key clinical message: Previous leptospirosis should be considered in a dog with calcinosis cutis. The cutaneous lesions appeared after acute leptospirosis and regressed spontaneously.


Calcinose cutanée localisée associée à une leptospirose chez un chiot Beagle de 4 mois. Un Beagle mâle de 4 mois a été présenté en consultation à la suite de l'apparition de nodules cutanés fermes 15 jours auparavant. L'examen histopathologique de biopsies cutanées a révélé une calcinose cutanée. Le contrôle à 40 jours a confirmé une résolution spontanée des lésions sans traitement spécifique. Deux semaines avant le développement des lésions cutanées, ce chien avait été hospitalisé et traité pour une maladie rénale et hépatique, attribuée à une leptospirose. Les examens PCR et sérologique étaient positifs pour Leptospira australis. La calcinose cutanée secondaire à une maladie systémique (leptospirose, blastomycose) est rarement rapportée et le mécanisme étiopathogénique suspecté comprenait un stress organique (hypersécrétion de cortisol) et un déséquilibre du métabolisme phosphocalcique lors de l'épisode d'insuffisance rénale aiguë. À notre connaissance, il s'agit du troisième cas publié de calcinose cutanée associée à la leptospirose chez le chien.Message clinique clé:Une potentielle leptospirose antérieure doit être mentionnée chez un chien atteint de calcinose cutanée. Les lésions cutanées semblent apparaître de manière décalée et régresser spontanément.(Traduit par Claude Muller).


Asunto(s)
Calcinosis , Enfermedades de los Perros , Leptospirosis , Neoplasias Cutáneas , Animales , Calcinosis/complicaciones , Calcinosis/veterinaria , Calcio , Enfermedades de los Perros/diagnóstico , Perros , Hidrocortisona , Leptospirosis/complicaciones , Leptospirosis/veterinaria , Masculino , Fósforo , Neoplasias Cutáneas/veterinaria
9.
Mol Oncol ; 16(17): 3167-3191, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35838338

RESUMEN

In glioblastoma (GBM), tumour-associated microglia/macrophages (TAMs) represent the major cell type of the stromal compartment and contribute to tumour immune escape mechanisms. Thus, targeting TAMs is emerging as a promising strategy for immunotherapy. However, TAM heterogeneity and metabolic adaptation along GBM progression represent critical features for the design of effective TAM-targeted therapies. Here, we comprehensively study the cellular and molecular changes of TAMs in the GL261 GBM mouse model, combining single-cell RNA-sequencing with flow cytometry and immunohistological analyses along GBM progression and in the absence of Acod1 (also known as Irg1), a key gene involved in the metabolic reprogramming of macrophages towards an anti-inflammatory phenotype. Similarly to patients, we identify distinct TAM profiles, mainly based on their ontogeny, that reiterate the idea that microglia- and macrophage-like cells show key transcriptional differences and dynamically adapt along GBM stages. Notably, we uncover decreased antigen-presenting cell features and immune reactivity in TAMs along tumour progression that are instead enhanced in Acod1-deficient mice. Overall, our results provide insight into TAM heterogeneity and highlight a novel role for Acod1 in TAM adaptation during GBM progression.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Neoplasias Encefálicas/patología , Glioblastoma/metabolismo , Inmunoterapia , Macrófagos/metabolismo , Ratones , Microglía/metabolismo , Microglía/patología , Microambiente Tumoral
10.
Proteomics Clin Appl ; 16(5): e2200015, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35579911

RESUMEN

PURPOSE: Parkinson's disease (PD) is the second most prevalent neurodegenerative disease. It is generally diagnosed clinically after the irreversible loss of dopaminergic neurons and no general biomarkers currently exist. To gain insight into the underlying cellular causes of PD we aimed to quantify the proteomic differences between healthy control and PD patient cells. EXPERIMENTAL DESIGN: Sequential Window Acquisition of all THeoretical Mass Spectra was performed on primary cells from healthy controls and PD patients. RESULTS: In total, 1948 proteins were quantified and 228 proteins were significantly differentially expressed in PD patient cells. In PD patient cells, we identified seven significantly increased proteins involved in the unfolded protein response (UPR) and focused on cells with high and low amounts of PDIA6 and HYOU1. We discovered that PD patients with high amounts of PDIA6 and HYOU1 proteins were more sensitive to endoplasmic reticulum stress, in particular to tunicamycin. Data is available via ProteomeXchange with identifier PXD030723. CONCLUSIONS AND CLINICAL RELEVANCE: This data from primary patient cells has uncovered a critical role of the UPR in patients with PD and may provide insight to the underlying cellular dysfunctions in these patients.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Biomarcadores , Humanos , Enfermedad de Parkinson/metabolismo , Proteómica , Tunicamicina/farmacología
11.
Nat Commun ; 11(1): 6366, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33311477

RESUMEN

The infiltrative nature of Glioblastoma (GBM), the most aggressive primary brain tumor, critically prevents complete surgical resection and masks tumor cells behind the blood brain barrier reducing the efficacy of systemic treatment. Here, we use a genome-wide interference screen to determine invasion-essential genes and identify the AN1/A20 zinc finger domain containing protein 3 (ZFAND3) as a crucial driver of GBM invasion. Using patient-derived cellular models, we show that loss of ZFAND3 hampers the invasive capacity of GBM, whereas ZFAND3 overexpression increases motility in cells that were initially not invasive. At the mechanistic level, we find that ZFAND3 activity requires nuclear localization and integral zinc-finger domains. Our findings indicate that ZFAND3 acts within a nuclear protein complex to activate gene transcription and regulates the promoter of invasion-related genes such as COL6A2, FN1, and NRCAM. Further investigation in ZFAND3 function in GBM and other invasive cancers is warranted.


Asunto(s)
Neoplasias Encefálicas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Glioblastoma/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Neoplasias Encefálicas/patología , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular , Colágeno Tipo VI/genética , Fibronectinas/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Ratones , Invasividad Neoplásica/genética , Dominios Proteicos , Transcriptoma
12.
Acta Neuropathol ; 140(6): 919-949, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33009951

RESUMEN

Patient-based cancer models are essential tools for studying tumor biology and for the assessment of drug responses in a translational context. We report the establishment a large cohort of unique organoids and patient-derived orthotopic xenografts (PDOX) of various glioma subtypes, including gliomas with mutations in IDH1, and paired longitudinal PDOX from primary and recurrent tumors of the same patient. We show that glioma PDOXs enable long-term propagation of patient tumors and represent clinically relevant patient avatars that retain histopathological, genetic, epigenetic, and transcriptomic features of parental tumors. We find no evidence of mouse-specific clonal evolution in glioma PDOXs. Our cohort captures individual molecular genotypes for precision medicine including mutations in IDH1, ATRX, TP53, MDM2/4, amplification of EGFR, PDGFRA, MET, CDK4/6, MDM2/4, and deletion of CDKN2A/B, PTCH, and PTEN. Matched longitudinal PDOX recapitulate the limited genetic evolution of gliomas observed in patients following treatment. At the histological level, we observe increased vascularization in the rat host as compared to mice. PDOX-derived standardized glioma organoids are amenable to high-throughput drug screens that can be validated in mice. We show clinically relevant responses to temozolomide (TMZ) and to targeted treatments, such as EGFR and CDK4/6 inhibitors in (epi)genetically defined subgroups, according to MGMT promoter and EGFR/CDK status, respectively. Dianhydrogalactitol (VAL-083), a promising bifunctional alkylating agent in the current clinical trial, displayed high therapeutic efficacy, and was able to overcome TMZ resistance in glioblastoma. Our work underscores the clinical relevance of glioma organoids and PDOX models for translational research and personalized treatment studies and represents a unique publicly available resource for precision oncology.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Xenoinjertos/inmunología , Organoides/patología , Temozolomida/uso terapéutico , Animales , Neoplasias Encefálicas/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioma/genética , Xenoinjertos/efectos de los fármacos , Humanos , Ratones , Recurrencia Local de Neoplasia/genética , Organoides/inmunología , Medicina de Precisión/métodos , Ratas
13.
Cancers (Basel) ; 12(9)2020 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-32927769

RESUMEN

Resistance to chemotherapy by temozolomide (TMZ) is a major cause of glioblastoma (GBM) recurrence. So far, attempts to characterize factors that contribute to TMZ sensitivity have largely focused on protein-coding genes, and failed to provide effective therapeutic targets. Long noncoding RNAs (lncRNAs) are essential regulators of epigenetic-driven cell diversification, yet, their contribution to the transcriptional response to drugs is less understood. Here, we performed RNA-seq and small RNA-seq to provide a comprehensive map of transcriptome regulation upon TMZ in patient-derived GBM stem-like cells displaying different drug sensitivity. In a search for regulatory mechanisms, we integrated thousands of molecular associations stored in public databases to generate a background "RNA interactome". Our systems-level analysis uncovered a coordinated program of TMZ response reflected by regulatory circuits that involve transcription factors, mRNAs, miRNAs, and lncRNAs. We discovered 22 lncRNAs involved in regulatory loops and/or with functional relevance in drug response and prognostic value in gliomas. Thus, the investigation of TMZ-induced gene networks highlights novel RNA-based predictors of chemosensitivity in GBM. The computational modeling used to identify regulatory circuits underlying drug response and prioritizing gene candidates for functional validation is applicable to other datasets.

14.
N Biotechnol ; 54: 52-61, 2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31398512

RESUMEN

Next-generation sequencing (NGS) analyses on DNA derived from archived Formalin-Fixed Paraffin-Embedded (FFPE) clinical material can provide a powerful tool in oncology research and clinical diagnostics. Although several studies have established that NGS can be performed using DNA from FFPE tissue, the accuracy and reproducibility of such analyses, as well as their robustness to the biomolecular quality of the samples used, remains a matter of debate. Excellent reviews have recently been published, providing evidence-based best practices for FFPE DNA extraction. Alternative fixatives exist, although their implementation in clinical practice is difficult. In this article, we present (i) a review of fixed tissue DNA preanalytics with a special focus on DNA extraction and fixed tissue sample qualification and (ii) results from comparisons between different methods of DNA extraction from tissue samples that have been fixed or stabilized by different methods, in terms of NGS metrics and different DNA quality metrics.


Asunto(s)
ADN/análisis , ADN/aislamiento & purificación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Fase Preanalítica/normas , Fijación del Tejido , ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Control de Calidad
15.
BMC Med Genomics ; 12(1): 132, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31533822

RESUMEN

BACKGROUND: The amount of publicly available cancer-related "omics" data is constantly growing and can potentially be used to gain insights into the tumour biology of new cancer patients, their diagnosis and suitable treatment options. However, the integration of different datasets is not straightforward and requires specialized approaches to deal with heterogeneity at technical and biological levels. METHODS: Here we present a method that can overcome technical biases, predict clinically relevant outcomes and identify tumour-related biological processes in patients using previously collected large discovery datasets. The approach is based on independent component analysis (ICA) - an unsupervised method of signal deconvolution. We developed parallel consensus ICA that robustly decomposes transcriptomics datasets into expression profiles with minimal mutual dependency. RESULTS: By applying the method to a small cohort of primary melanoma and control samples combined with a large discovery melanoma dataset, we demonstrate that our method distinguishes cell-type specific signals from technical biases and allows to predict clinically relevant patient characteristics. We showed the potential of the method to predict cancer subtypes and estimate the activity of key tumour-related processes such as immune response, angiogenesis and cell proliferation. ICA-based risk score was proposed and its connection to patient survival was validated with an independent cohort of patients. Additionally, through integration of components identified for mRNA and miRNA data, the proposed method helped deducing biological functions of miRNAs, which would otherwise not be possible. CONCLUSIONS: We present a method that can be used to map new transcriptomic data from cancer patient samples onto large discovery datasets. The method corrects technical biases, helps characterizing activity of biological processes or cell types in the new samples and provides the prognosis of patient survival.


Asunto(s)
Biología Computacional/métodos , Melanoma/genética , MicroARNs/metabolismo , Transcriptoma , Bases de Datos Genéticas , Femenino , Humanos , Masculino , Melanoma/mortalidad , Melanoma/patología , MicroARNs/genética , Análisis de Componente Principal , Análisis de Supervivencia
16.
Neuro Oncol ; 21(7): 890-900, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-30958558

RESUMEN

BACKGROUND: Suicide gene therapy for malignant gliomas has shown encouraging results in the latest clinical trials. However, prodrug application was most often restricted to short-term treatment (14 days), especially when replication-defective vectors were used. We previously showed that a substantial fraction of herpes simplex virus thymidine kinase (HSV-TK) transduced tumor cells survive ganciclovir (GCV) treatment in an orthotopic glioblastoma (GBM) xenograft model. Here we analyzed whether these TK+ tumor cells are still sensitive to prodrug treatment and whether prolonged prodrug treatment can enhance treatment efficacy. METHODS: Glioma cells positive for TK and green fluorescent protein (GFP) were sorted from xenograft tumors recurring after suicide gene therapy, and their sensitivity to GCV was tested in vitro. GBM xenografts were treated with HSV-TK/GCV, HSV-TK/valganciclovir (valGCV), or HSV-TK/valGCV + erlotinib. Tumor growth was analyzed by MRI, and survival as well as morphological and molecular changes were assessed. RESULTS: TK-GFP+ tumor cells from recurrent xenograft tumors retained sensitivity to GCV in vitro. Importantly, a prolonged period (3 mo) of prodrug administration with valganciclovir (valGCV) resulted in a significant survival advantage compared with short-term (3 wk) application of GCV. Recurrent tumors from the treatment groups were more invasive and less angiogenic compared with primary tumors and showed significant upregulation of epidermal growth factor receptor (EGFR) expression. However, double treatment with the EGFR inhibitor erlotinib did not increase therapeutic efficacy. CONCLUSION: Long-term treatment with valGCV should be considered as a replacement for short-term treatment with GCV in clinical trials of HSV-TK mediated suicide gene therapy.


Asunto(s)
Antivirales/farmacología , Terapia Genética , Glioblastoma/terapia , Profármacos/farmacología , Timidina Quinasa/genética , Valganciclovir/farmacología , Adenoviridae/genética , Animales , Apoptosis , Proliferación Celular , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Invasividad Neoplásica , Simplexvirus/enzimología , Timidina Quinasa/administración & dosificación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Nat Commun ; 10(1): 1787, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992437

RESUMEN

The identity and unique capacity of cancer stem cells (CSC) to drive tumor growth and resistance have been challenged in brain tumors. Here we report that cells expressing CSC-associated cell membrane markers in Glioblastoma (GBM) do not represent a clonal entity defined by distinct functional properties and transcriptomic profiles, but rather a plastic state that most cancer cells can adopt. We show that phenotypic heterogeneity arises from non-hierarchical, reversible state transitions, instructed by the microenvironment and is predictable by mathematical modeling. Although functional stem cell properties were similar in vitro, accelerated reconstitution of heterogeneity provides a growth advantage in vivo, suggesting that tumorigenic potential is linked to intrinsic plasticity rather than CSC multipotency. The capacity of any given cancer cell to reconstitute tumor heterogeneity cautions against therapies targeting CSC-associated membrane epitopes. Instead inherent cancer cell plasticity emerges as a novel relevant target for treatment.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/genética , Plasticidad de la Célula/efectos de los fármacos , Glioblastoma/genética , Microambiente Tumoral/efectos de los fármacos , Animales , Antineoplásicos Alquilantes/uso terapéutico , Biopsia , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Plasticidad de la Célula/genética , Estudios de Cohortes , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Temozolomida/farmacología , Temozolomida/uso terapéutico , Resultado del Tratamiento , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
iScience ; 13: 351-370, 2019 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-30884312

RESUMEN

CCAAT enhancer-binding protein beta (C/EBPß) is a pioneer transcription factor that specifies cell differentiation. C/EBPß is intrinsically unstructured, a molecular feature common to many proteins involved in signal processing and epigenetics. The structure of C/EBPß differs depending on alternative translation initiation and multiple post-translational modifications (PTM). Mutation of distinct PTM sites in C/EBPß alters protein interactions and cell differentiation, suggesting that a C/EBPß PTM indexing code determines epigenetic outcomes. Herein, we systematically explored the interactome of C/EBPß using an array technique based on spot-synthesized C/EBPß-derived linear tiling peptides with and without PTM, combined with mass spectrometric proteomic analysis of protein interactions. We identified interaction footprints of ∼1,300 proteins in nuclear extracts, many with chromatin modifying, chromatin remodeling, and RNA processing functions. The results suggest that C/EBPß acts as a multi-tasking molecular switchboard, integrating signal-dependent modifications and structural plasticity to orchestrate interactions with numerous protein complexes directing cell fate and function.

19.
Brain Pathol ; 29(4): 513-529, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30506802

RESUMEN

While the central nervous system is considered an immunoprivileged site and brain tumors display immunosuppressive features, both innate and adaptive immune responses affect glioblastoma (GBM) growth and treatment resistance. However, the impact of the major immune cell population in gliomas, represented by glioma-associated microglia/macrophages (GAMs), on patients' clinical course is still unclear. Thus, we aimed at assessing the immunohistochemical expression of selected microglia and macrophage markers in 344 gliomas (including gliomas from WHO grade I-IV). Furthermore, we analyzed a cohort of 241 IDH1R132H-non-mutant GBM patients for association of GAM subtypes and patient overall survival. Phenotypical properties of GAMs, isolated from high-grade astrocytomas by CD11b-based magnetic cell sorting, were analyzed by immunocytochemistry, mRNA microarray, qRT-PCR and bioinformatic analyses. A higher amount of CD68-, CD163- and CD206-positive GAMs in the vital tumor core was associated with beneficial patient survival. The mRNA expression profile of GAMs displayed an upregulation of factors that are considered as pro-inflammatory M1 (eg, CCL2, CCL3L3, CCL4, PTGS2) and anti-inflammatory M2 polarization markers (eg, MRC1, LGMN, CD163, IL10, MSR1), the latter rather being associated with phagocytic functions in the GBM microenvironment. In summary, we present evidence that human GBMs contain mixed M1/M2-like polarized GAMs and that the levels of different GAM subpopulations in the tumor core are positively associated with overall survival of patients with IDH1R132H-non-mutant GBMs.


Asunto(s)
Glioma/patología , Macrófagos/patología , Microglía/patología , Adulto , Anciano , Animales , Astrocitoma/patología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Femenino , Glioblastoma/patología , Glioma/inmunología , Glioma/metabolismo , Humanos , Inmunohistoquímica , Macrófagos/inmunología , Masculino , Ratones , Microglía/inmunología , Persona de Mediana Edad , Pronóstico , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Surg Laparosc Endosc Percutan Tech ; 28(4): 256-260, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29965896

RESUMEN

BACKGROUND: Percutaneous cholecystostomy (PC) is an alternative to cholecystectomy for acute calculous cholecystitis (ACC) in patients with high mortality risk, but the catheters' and patients' management remains unclear. This study aimed to determine outcomes after PC and to define surgical strategy. METHODS: All patients who underwent PC between 2009 and 2014 for ACC in a single institution were reviewed for outcomes and postdrainage management. RESULTS: Forty-one patients underwent PC with a median age of 77 years. Twenty patients (45%) presented American Society of Anesthesiologists (ASA) score 3-4, and all had cholecystitis grade II or III. The 6-month mortality was 17% (7/41 patients). Twelve patients (29%) presented PC-related complications. Catheters were removed, clamped, left open, and dislodged for 17, 9, 5, and 5 patients, respectively. Three patients died after PC, and data were missing for 2 patients. Twenty-five (61%) patients underwent cholecystectomy after PC, and were significantly younger (median age, 70 vs. 83; P<0.010), and presented with higher ASA score (ASA: 3-4, 36% vs. 68%; P=0.041) and more grade III cholecystitis (16% vs. 44%; P=0.050). Among 16 patients who did not undergo cholecystectomy, none presented recurrent gallstone disease (median survival, 7 mo; range, 0 to 55). CONCLUSIONS: The risk of complications associated with the catheter is high. To prevent morbidity related to PC, we suggest removing the catheter when ACC is controlled.


Asunto(s)
Colecistectomía Laparoscópica/instrumentación , Colecistitis/cirugía , Anciano , Anciano de 80 o más Años , Cateterismo/efectos adversos , Cateterismo/instrumentación , Catéteres de Permanencia/efectos adversos , Colecistectomía Laparoscópica/efectos adversos , Colecistectomía Laparoscópica/métodos , Drenaje/efectos adversos , Drenaje/instrumentación , Estudios de Factibilidad , Femenino , Cálculos Biliares/cirugía , Humanos , Masculino , Persona de Mediana Edad , Cuidados Posoperatorios/métodos , Complicaciones Posoperatorias/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA