Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuro Oncol ; 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38607967

RESUMO

BACKGROUND: With the recognition that noncancerous cells function as critical regulators of brain tumor growth, we recently demonstrated that neurons drive low-grade glioma initiation and progression. Using mouse models of neurofibromatosis type 1 (NF1)-associated optic pathway glioma (OPG), we showed that Nf1 mutation induces neuronal hyperexcitability and midkine expression, which activates an immune axis to support tumor growth, such that high-dose lamotrigine treatment reduces Nf1-OPG proliferation. Herein, we execute a series of complementary experiments to address several key knowledge gaps relevant to future clinical translation. METHODS: We leverage a collection of Nf1-mutant mice that spontaneously develop OPGs to alter both germline and retinal neuron-specific midkine expression. Nf1-mutant mice harboring several different NF1 patient-derived germline mutations were employed to evaluate neuronal excitability and midkine expression. Two distinct Nf1-OPG preclinical mouse models were used to assess lamotrigine effects on tumor progression and growth in vivo. RESULTS: We establish that neuronal midkine is both necessary and sufficient for Nf1-OPG growth, demonstrating an obligate relationship between germline Nf1 mutation, neuronal excitability, midkine production, and Nf1-OPG proliferation. We show anti-epileptic drug (lamotrigine) specificity in suppressing neuronal midkine production. Relevant to clinical translation, lamotrigine prevents Nf1-OPG progression and suppresses the growth of existing tumors for months following drug cessation. Importantly, lamotrigine abrogates tumor growth in two Nf1-OPG strains using pediatric epilepsy clinical dosing. CONCLUSIONS: Together, these findings establish midkine and neuronal hyperexcitability as targetable drivers of Nf1-OPG growth and support the use of lamotrigine as a potential chemoprevention or chemotherapy agent for children with NF1-OPG.

2.
Acta Neuropathol Commun ; 12(1): 21, 2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38308315

RESUMO

Tissue injury and tumorigenesis share many cellular and molecular features, including immune cell (T cells, monocytes) infiltration and inflammatory factor (cytokines, chemokines) elaboration. Their common pathobiology raises the intriguing possibility that brain injury could create a tissue microenvironment permissive for tumor formation. Leveraging several murine models of the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome and two experimental methods of brain injury, we demonstrate that both optic nerve crush and diffuse traumatic brain injury induce optic glioma (OPG) formation in mice harboring Nf1-deficient preneoplastic progenitors. We further elucidate the underlying molecular and cellular mechanisms, whereby glutamate released from damaged neurons stimulates IL-1ß release by oligodendrocytes to induce microglia expression of Ccl5, a growth factor critical for Nf1-OPG formation. Interruption of this cellular circuit using glutamate receptor, IL-1ß or Ccl5 inhibitors abrogates injury-induced glioma progression, thus establishing a causative relationship between injury and tumorigenesis.


Assuntos
Lesões Encefálicas , Neurofibromatose 1 , Glioma do Nervo Óptico , Camundongos , Animais , Glioma do Nervo Óptico/metabolismo , Glioma do Nervo Óptico/patologia , Neurofibromatose 1/patologia , Microglia/metabolismo , Lesões Encefálicas/metabolismo , Neurônios/metabolismo , Carcinogênese/metabolismo , Microambiente Tumoral
3.
Ann Clin Transl Neurol ; 11(3): 812-818, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38229454

RESUMO

Optic pathway gliomas (OPGs) arising in children with neurofibromatosis type 1 (NF1) can cause retinal ganglion cell (RGC) dysfunction and vision loss, which occurs more frequently in girls. While our previous studies demonstrated that estrogen was partly responsible for this sexually dimorphic visual impairment, herein we elucidate the underlying mechanism. In contrast to their male counterparts, female Nf1OPG mice have increased expression of glial interleukin-1ß (IL-1ß), which is neurotoxic to RGCs in vitro. Importantly, both IL-1ß neutralization and leuprolide-mediated estrogen suppression decrease IL-1ß expression and ameliorate RGC dysfunction, providing preclinical proof-of-concept evidence supporting novel neuroprotective strategies for NF1-OPG-induced vision loss.


Assuntos
Neurofibromatose 1 , Glioma do Nervo Óptico , Humanos , Criança , Masculino , Feminino , Camundongos , Animais , Glioma do Nervo Óptico/metabolismo , Células Ganglionares da Retina , Interleucina-1beta/metabolismo , Transtornos da Visão , Estrogênios/farmacologia , Estrogênios/metabolismo
4.
Dis Model Mech ; 16(12)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37990867

RESUMO

Neurofibromatosis type 1 (NF1) is an autosomal dominant condition caused by germline mutations in the neurofibromin 1 (NF1) gene. Children with NF1 are prone to the development of multiple nervous system abnormalities, including autism and brain tumors, which could reflect the effect of NF1 mutation on microglia function. Using heterozygous Nf1-mutant mice, we previously demonstrated that impaired purinergic signaling underlies deficits in microglia process extension and phagocytosis in situ. To determine whether these abnormalities are also observed in human microglia in the setting of NF1, we leveraged an engineered isogenic series of human induced pluripotent stem cells to generate human microglia-like (hiMGL) cells heterozygous for three different NF1 gene mutations found in patients with NF1. Whereas all NF1-mutant and isogenic control hiMGL cells expressed classical microglia markers and exhibited similar transcriptomes and cytokine/chemokine release profiles, only NF1-mutant hiMGL cells had defects in P2X receptor activation, phagocytosis and motility. Taken together, these findings indicate that heterozygous NF1 mutations impair a subset of the functional properties of human microglia, which could contribute to the neurological abnormalities seen in children with NF1.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neurofibromatose 1 , Animais , Humanos , Camundongos , Genes da Neurofibromatose 1 , Microglia/patologia , Mutação/genética , Neurofibromatose 1/genética , Neurofibromina 1/genética
5.
Acta Neuropathol Commun ; 10(1): 120, 2022 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-35986378

RESUMO

A major obstacle to identifying improved treatments for pediatric low-grade brain tumors (gliomas) is the inability to reproducibly generate human xenografts. To surmount this barrier, we leveraged human induced pluripotent stem cell (hiPSC) engineering to generate low-grade gliomas (LGGs) harboring the two most common pediatric pilocytic astrocytoma-associated molecular alterations, NF1 loss and KIAA1549:BRAF fusion. Herein, we identified that hiPSC-derived neuroglial progenitor populations (neural progenitors, glial restricted progenitors and oligodendrocyte progenitors), but not terminally differentiated astrocytes, give rise to tumors retaining LGG histologic features for at least 6 months in vivo. Additionally, we demonstrated that hiPSC-LGG xenograft formation requires the absence of CD4 T cell-mediated induction of astrocytic Cxcl10 expression. Genetic Cxcl10 ablation is both necessary and sufficient for human LGG xenograft development, which additionally enables the successful long-term growth of patient-derived pediatric LGGs in vivo. Lastly, MEK inhibitor (PD0325901) treatment increased hiPSC-LGG cell apoptosis and reduced proliferation both in vitro and in vivo. Collectively, this study establishes a tractable experimental humanized platform to elucidate the pathogenesis of and potential therapeutic opportunities for childhood brain tumors.


Assuntos
Astrocitoma , Neoplasias Encefálicas , Glioma , Células-Tronco Pluripotentes Induzidas , Animais , Astrocitoma/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Criança , Glioma/genética , Glioma/metabolismo , Glioma/terapia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Neuroglia/patologia
6.
Nat Commun ; 13(1): 2785, 2022 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-35589737

RESUMO

Neuronal activity is emerging as a driver of central and peripheral nervous system cancers. Here, we examined neuronal physiology in mouse models of the tumor predisposition syndrome Neurofibromatosis-1 (NF1), with different propensities to develop nervous system cancers. We show that central and peripheral nervous system neurons from mice with tumor-causing Nf1 gene mutations exhibit hyperexcitability and increased secretion of activity-dependent tumor-promoting paracrine factors. We discovered a neurofibroma mitogen (COL1A2) produced by peripheral neurons in an activity-regulated manner, which increases NF1-deficient Schwann cell proliferation, establishing that neurofibromas are regulated by neuronal activity. In contrast, mice with the Arg1809Cys Nf1 mutation, found in NF1 patients lacking neurofibromas or optic gliomas, do not exhibit neuronal hyperexcitability or develop these NF1-associated tumors. The hyperexcitability of tumor-prone Nf1-mutant neurons results from reduced NF1-regulated hyperpolarization-activated cyclic nucleotide-gated (HCN) channel function, such that neuronal excitability, activity-regulated paracrine factor production, and tumor progression are attenuated by HCN channel activation. Collectively, these findings reveal that NF1 mutations act at the level of neurons to modify tumor predisposition by increasing neuronal excitability and activity-regulated paracrine factor production.


Assuntos
Neurofibroma , Neurofibromatose 1 , Glioma do Nervo Óptico , Animais , Humanos , Camundongos , Neurofibroma/patologia , Neurofibromatose 1/genética , Neurofibromina 1/genética , Neurônios/patologia , Glioma do Nervo Óptico/patologia , Sistema Nervoso Periférico/patologia , Células de Schwann/patologia
7.
Neurooncol Adv ; 4(1): vdab194, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35187488

RESUMO

BACKGROUND: Brain tumor formation and progression are dictated by cooperative interactions between neoplastic and non-neoplastic cells. This stromal dependence is nicely illustrated by tumors arising in the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome, where children develop low-grade optic pathway gliomas (OPGs). Using several authenticated Nf1-OPG murine models, we previously demonstrated that murine Nf1-OPG growth is regulated by T cell function and microglia Ccl5 production, such that their inhibition reduces tumor proliferation in vivo. While these interactions are critical for established Nf1-OPG tumor growth, their importance in tumor formation has not been explored. METHODS: A combination of bulk and single-cell RNA mouse optic nerve sequencing, immunohistochemistry, T cell assays, and pharmacologic and antibody-mediated inhibition methods were used in these experiments. RESULTS: We show that T cells and microglia are the main non-neoplastic immune cell populations in both murine and human LGGs. Moreover, we demonstrate that CD8+ T cells, the predominant LGG-infiltrating lymphocyte population, are selectively recruited through increased Ccl2 receptor (Ccr4) expression in CD8+, but not CD4+, T cells, in a NF1/RAS-dependent manner. Finally, we identify the times during gliomagenesis when microglia Ccl5 production (3-6 weeks of age) and Ccl2-mediated T cell infiltration (7-10 weeks of age) occur, such that temporally-restricted Ccl2 or Ccl5 inhibition abrogates tumor formation >3.5 months following the cessation of treatment. CONCLUSIONS: Collectively, these findings provide proof-of-concept demonstrations that targeting stromal support during early gliomagenesis durably blocks murine LGG formation.

8.
Neuro Oncol ; 24(1): 14-26, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34043012

RESUMO

BACKGROUND: Emerging insights from numerous laboratories have revealed important roles for nonneoplastic cells in the development and progression of brain tumors. One of these nonneoplastic cellular constituents, glioma-associated microglia (GAM), represents a unique population of brain monocytes within the tumor microenvironment that have been reported to both promote and inhibit glioma proliferation. To elucidate the role of GAM in the setting of low-grade glioma (LGG), we leveraged RNA sequencing meta-analysis, genetically engineered mouse strains, and human biospecimens. METHODS: Publicly available disease-associated microglia (DAM) RNA-seq datasets were used, followed by immunohistochemistry and RNAScope validation. CD11a-deficient mouse microglia were used for in vitro functional studies, while LGG growth in mice was assessed using anti-CD11a neutralizing antibody treatment of Neurofibromatosis type 1 (Nf1) optic glioma mice in vivo. RESULTS: We identified Itgal/CD11a enrichment in GAM relative to other DAM populations, which was confirmed in several independently generated murine models of Nf1 optic glioma. Moreover, ITGAL/CD11A expression was similarly increased in human LGG (pilocytic astrocytoma) specimens from several different datasets, specifically in microglia from these tumors. Using CD11a-knockout mice, CD11a expression was shown to be critical for murine microglia CX3CL1 receptor (Cx3cr1) expression and CX3CL1-directed motility, as well as glioma mitogen (Ccl5) production. Consistent with an instructive role for CD11a+ microglia in stromal control of LGG growth, antibody-mediated CD11a inhibition reduced mouse Nf1 LGG growth in vivo. CONCLUSIONS: Collectively, these findings establish ITGAL/CD11A as a critical microglia regulator of LGG biology relevant to future stroma-targeted brain tumor treatment strategies.


Assuntos
Neoplasias Encefálicas , Neurofibromatose 1 , Glioma do Nervo Óptico , Animais , Neoplasias Encefálicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Microglia , Análise de Sequência de RNA , Microambiente Tumoral
9.
Nat Commun ; 12(1): 7122, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34880260

RESUMO

To elucidate the mechanisms underlying the reduced incidence of brain tumors in children with Neurofibromatosis type 1 (NF1) and asthma, we leverage Nf1 optic pathway glioma (Nf1OPG) mice, human and mouse RNAseq data, and two different experimental asthma models. Following ovalbumin or house dust mite asthma induction at 4-6 weeks of age (WOA), Nf1OPG mouse optic nerve volumes and proliferation are decreased at 12 and 24 WOA, indicating no tumor development. This inhibition is accompanied by reduced expression of the microglia-produced optic glioma mitogen, Ccl5. Human and murine T cell transcriptome analyses reveal that inhibition of microglia Ccl5 production results from increased T cell expression of decorin, which blocks Ccl4-mediated microglia Ccl5 expression through reduced microglia NFκB signaling. Decorin or NFκB inhibitor treatment of Nf1OPG mice at 4-6 WOA inhibits tumor formation at 12 WOA, thus establishing a potential mechanistic etiology for the attenuated glioma incidence observed in children with asthma.


Assuntos
Asma/imunologia , Asma/metabolismo , Decorina/metabolismo , Glioma , Microglia/metabolismo , Linfócitos T/imunologia , Animais , Neoplasias Encefálicas/patologia , Quimiocina CCL4/metabolismo , Quimiocina CCL5/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Monitorização Imunológica , Neurofibromatose 1/metabolismo , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Nervo Óptico/metabolismo , Glioma do Nervo Óptico/patologia , Transdução de Sinais
10.
J Biochem Mol Toxicol ; 33(2): e22242, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30368985

RESUMO

The endoplasmic reticulum (ER) plays an important role in the regulation and maintenance of cellular homeostasis. However, unresolved ER stress leads to deleterious effects by inducing the accumulation of unfolded proteins in the cell. Here we have demonstrated the protective aspects of quercetin against radiation-induced ER stress and against inflammation in primary cultured dorsal root ganglion (DRG) neurons. The mature DRG neurons were pretreated with different concentrations of quercetin (5-100 µM) for 24 hours before 2 Gy gamma radiation exposure and then subjected to a cytotoxicity assay, quantitative real-time polymerase chain reaction and Western blot analysis. The results showed that quercetin decreased the expression of BiP and C/EBP-homologous protein, the ER stress marker genes along with downregulation of tumor necrosis factor-α, JNK in irradiated DRG neurons. Furthermore, quercetin pretreatment significantly increased the cytoskeletal protein Tuj1 and the neurotrophin brain-derived neurotrophic factor in the neuron. These results indicate that quercetin plays a neuroprotective role against radiation-mediated ER stress and inflammatory responses.


Assuntos
Estresse do Retículo Endoplasmático , Raios gama/efeitos adversos , Gânglios Espinais/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Quercetina/farmacologia , Animais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos da radiação , Gânglios Espinais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/patologia
11.
Pharm Nanotechnol ; 6(3): 201-208, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29886841

RESUMO

BACKGROUND: Facile, environmental friendly synthesis of metal oxide nanoparticles is of paramount importance when its biological applications are considered. OBJECTIVE: Current study reports phyto-fabrication of Zinc oxide nanoparticles using aqueous extract of Piper betel leaves as stabilizing and capping agent using co-precipitation method. RESULTS: P betel synthesized ZnO nanoparticles (PZnO) was characterized using Powder X-ray diffraction, UV-Visible spectroscopy, scanning electron microscopy and dynamic light scattering. PXRD pattern demonstrated hexagonal wurtzite structure with preferred orientation (100) plane confirmed by JCPDS file 36-1451. UV- Vis spectroscopy showed peak at 370 nm due to band edge of semiconductor, the PZnO. The average particle size determined by DLS measurement was 69 nm and morphologically the particles were short rod shaped and agglomerated as demonstrated by SEM images. Antibacterial activity of PZnO against dental pathogens namely Streptococcus mutans and Lactobacillus acidphillus was performed using well-diffusion method and antioxidant activity against 2, 2 diphenyl 1 picrylhydrazyl (DPPH) free radicals and were compared with ZnO used in clinical dentistry (DZnO). PZnO showed higher antioxidant activity of ~70% at 200 µg/mL with consistent activity at lower aliquots. PZnO showed higher antimicrobial activity than DZnO against both tested microbes and also exhibited inhibitory effects in concentration as low as 3.25 µg/mL. Cytotoxicity study using Balb 3T3 mouse fibroblast cell lines showed <40% cellular growth inhibition by both PZnO and DZnO, indicating their benignity towards selected cell lines. CONCLUSION: Phyto-fabricated facile PZnO nanoparticles having demonstrable antibacterial and antioxidant activity can be considered for use in clinical dentistry after further clinical trials.


Assuntos
Antibacterianos , Antioxidantes , Nanopartículas Metálicas , Piper , Extratos Vegetais , Óxido de Zinco , Animais , Antibacterianos/administração & dosagem , Antibacterianos/química , Antioxidantes/administração & dosagem , Antioxidantes/química , Células 3T3 BALB , Compostos de Bifenilo/química , Sobrevivência Celular/efeitos dos fármacos , Odontologia , Lactobacillus acidophilus/efeitos dos fármacos , Lactobacillus acidophilus/crescimento & desenvolvimento , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Camundongos , Picratos/química , Extratos Vegetais/administração & dosagem , Extratos Vegetais/química , Folhas de Planta/química , Streptococcus mutans/efeitos dos fármacos , Streptococcus mutans/crescimento & desenvolvimento , Óxido de Zinco/administração & dosagem , Óxido de Zinco/química
12.
Metab Brain Dis ; 33(3): 855-868, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29429012

RESUMO

Ionizing radiation induces various pathophysiological conditions by altering central nervous system (CNS) homeostasis, leading to neurodegenerative diseases. However, the potential effect of ionizing radiation response on cellular physiology in glial cells is unclear. In the present study, micronucleus test, comet assay, and RT-PCR were performed to investigate the potential effect of gamma radiation in cultured oligodendrocytes and astrocytes with respect to genomic instability, Endoplasmic Reticulum (ER) stress, and inflammation. Further, we studied the effect of alteration in ER stress specific gene expression in cortex post whole body radiation in mice. Results showed that exposure of gamma radiation of 2Gy in-vitro cultured astrocytes and oligodendrocytes and 7Gy in-vivo induced ER stress and Inflammation along with profuse DNA damage and Chromosomal abnormality. Additionally, we observed downregulation of myelin basic protein levels in cultured oligodendrocytes exposed to radiation. The present data suggests that ER stress and pro inflammatory cytokines serve as the major players in inducing glial cell dysfunction post gamma irradiation along with induction of genomic instability. Taken together, these results indicate that ER stress, DNA damage, and inflammatory pathways may be critical events leading to glial cell dysfunction and subsequent cell death following exposure to ionizing radiation.


Assuntos
Astrócitos/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Instabilidade Genômica/genética , Neuroglia/metabolismo , Oligodendroglia/metabolismo , Animais , Morte Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...