Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Chem Phys ; 160(23)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38884404

RESUMO

Zinc tungstate is a semiconductor known for its favorable photocatalytic, photoluminescence, and scintillation properties, coupled with its relatively low cost, reduced toxicity, and high stability in biological and catalytic environments. In particular, zinc tungstate evinces scintillation properties, namely the ability to emit visible light upon absorption of energetic radiation such as x rays, which has led to applications not only as radiation detectors but also for biomedical applications involving the delivery of optical light to deep tissue, such as photodynamic therapy and optogenetics. Here, we report on the synthesis of zinc tungstate nanorods generated via an optimized but facile method, which allows for synthetic control over the aspect ratio of the as-synthesized anisotropic motifs via rational variation of the solution pH. We investigate the effect of aspect ratio on their resulting photoluminescent and radioluminescent properties. We further demonstrate the potential of these zinc tungstate nanorods for biomedical applications, such as photodynamic therapy for cancer treatment, by analyzing their toxicological profile within cell lines and neurons.


Assuntos
Nanotubos , Compostos de Tungstênio , Compostos de Tungstênio/química , Compostos de Tungstênio/toxicidade , Nanotubos/química , Humanos , Animais , Fotoquimioterapia , Sobrevivência Celular/efeitos dos fármacos , Compostos de Zinco/química , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Zinco/química
2.
J Pharmacol Exp Ther ; 389(1): 51-60, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38296645

RESUMO

Glioblastoma (GBM) is the most frequently diagnosed primary central nervous system tumor in adults. Despite the standard of care therapy, which includes surgical resection, temozolomide chemotherapy, radiation and the newly added tumor-treating fields, median survival remains only ∼20 months. Unfortunately, GBM has a ∼100% recurrence rate, but after recurrence there are no Food and Drug Administration-approved therapies to limit tumor growth and enhance patient survival, as these tumors are resistant to temozolomide (TMZ). Recently, our laboratory reported that lucanthone slows GBM by inhibiting autophagic flux through lysosome targeting and decreases the number of Olig2+ glioma stem-like cells (GSC) in vitro and in vivo. We now additionally report that lucanthone efficiently abates stemness in patient-derived GSC and reduces tumor microtube formation in GSC, an emerging hallmark of treatment resistance in GBM. In glioma tumors derived from cells with acquired resistance to TMZ, lucanthone retains the ability to perturb tumor growth, inhibits autophagy by targeting lysosomes, and reduces Olig2 positivity. We also find that lucanthone may act as an inhibitor of palmitoyl protein thioesterase 1. Our results suggest that lucanthone may function as a potential treatment option for GBM tumors that are not amenable to TMZ treatment. SIGNIFICANCE STATEMENT: We report that the antischistosome agent lucanthone impedes tumor growth in a preclinical model of temozolomide-resistant glioblastoma and reduces the numbers of stem-like glioma cells. In addition, it acts as an autophagy inhibitor, and its mechanism of action may be via inhibition of palmitoyl protein thioesterase 1. As there are no defined therapies approved for recurrent, TMZ-resistant tumor, lucanthone could emerge as a treatment for glioblastoma tumors that may not be amenable to TMZ both in the newly diagnosed and recurrent settings.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Lucantona , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Lucantona/farmacologia , Lucantona/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Proteínas de Membrana , Tioléster Hidrolases
3.
Glia ; 72(1): 111-132, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37675659

RESUMO

Chronic environmental stress and traumatic social experiences induce maladaptive behavioral changes and is a risk factor for major depressive disorder (MDD) and various anxiety-related psychiatric disorders. Clinical studies and animal models of chronic stress have reported that symptom severity is correlated with innate immune responses and upregulation of neuroinflammatory cytokine signaling in brain areas implicated in mood regulation (mPFC; medial Prefrontal Cortex). Despite increasing evidence implicating impairments of neuroplasticity and synaptic signaling deficits into the pathophysiology of stress-related mental disorders, how microglia may modulate neuronal homeostasis in response to chronic stress has not been defined. Here, using the repeated social defeat stress (RSDS) mouse model we demonstrate that microglial-induced inflammatory responses are regulating neuronal plasticity associated with psychosocial stress. Specifically, we show that chronic stress induces a rapid activation and proliferation of microglia as well as macrophage infiltration in the mPFC, and these processes are spatially related to neuronal activation. Moreover, we report a significant association of microglial inflammatory responses with susceptibility or resilience to chronic stress. In addition, we find that exposure to chronic stress exacerbates phagocytosis of synaptic elements and deficits in neuronal plasticity. Importantly, by utilizing two different CSF1R inhibitors (the brain penetrant PLX5622 and the non-penetrant PLX73086) we highlight a crucial role for microglia (and secondarily macrophages) in catalyzing the pathological manifestations linked to psychosocial stress in the mPFC and the resulting behavioral deficits usually associated with depression.


Assuntos
Transtorno Depressivo Maior , Microglia , Camundongos , Animais , Humanos , Microglia/patologia , Macrófagos , Neurônios , Estresse Psicológico/complicações , Estresse Psicológico/patologia
4.
Front Immunol ; 13: 958620, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36203599

RESUMO

In the past several years there has been a marked increase in our understanding of the pathophysiological hallmarks of glioblastoma development and progression, with specific respect to the contribution of the glioma tumor microenvironment to the rapid progression and treatment resistance of high-grade gliomas. Despite these strides, standard of care therapy still only targets rapidly dividing tumor cells in the glioma, and does little to curb the pro-tumorigenic functions of non-cancerous cells entrenched in the glioma microenvironment. This tumor promoting environment as well as the heterogeneity of high-grade gliomas contribute to the poor prognosis of this malignancy. The interaction of non-malignant cells in the microenvironment with the tumor cells accentuate phenotypes such as rapid proliferation or immunosuppression, so therapeutically modulating one target expressed on one cell type may be insufficient to restrain these rapidly developing neoplasias. With this in mind, identifying a target expressed on multiple cell types and understanding how it governs tumor-promoting functions in each cell type may have great utility in better managing this disease. Herein, we review the physiology and pathological effects of Neuropilin-1, a transmembrane co-receptor which mediates signal transduction pathways when associated with multiple other receptors. We discuss its effects on the properties of endothelial cells and on immune cell types within gliomas including glioma-associated macrophages, microglia, cytotoxic T cells and T regulatory cells. We also consider its effects when elaborated on the surface of tumor cells with respect to proliferation, stemness and treatment resistance, and review attempts to target Neuroplin-1 in the clinical setting.


Assuntos
Glioma , Neuropilina-1 , Células Endoteliais/metabolismo , Glioma/genética , Humanos , Microglia , Neuropilina-1/metabolismo , Microambiente Tumoral
5.
Front Oncol ; 12: 852940, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35494072

RESUMO

Glioblastoma is the most common and aggressive primary brain tumor in adults. Median survival time remains at 16-20 months despite multimodal treatment with surgical resection, radiation, temozolomide and tumor-treating fields therapy. After genotoxic stress glioma cells initiate cytoprotective autophagy, which contributes to treatment resistance, limiting the efficacy of these therapies and providing an avenue for glioma recurrence. Antagonism of autophagy steps has recently gained attention as it may enhance the efficacy of classical chemotherapies and newer immune-stimulating therapies. The modulation of autophagy in the clinic is limited by the low potency of common autophagy inhibitors and the inability of newer ones to cross the blood-brain barrier. Herein, we leverage lucanthone, an anti-schistosomal agent which crosses the blood-brain barrier and was recently reported to act as an autophagy inhibitor in breast cancer cells. Our studies show that lucanthone was toxic to glioma cells by inhibiting autophagy. It enhanced anti-glioma temozolomide (TMZ) efficacy at sub-cytotoxic concentrations, and suppressed the growth of stem-like glioma cells and temozolomide-resistant glioma stem cells. In vivo lucanthone slowed tumor growth: reduced numbers of Olig2+ glioma cells, normalized tumor vasculature, and reduced tumor hypoxia. We propose that lucanthone may serve to perturb a mechanism of temozolomide resistance and allow for successful treatment of TMZ-resistant glioblastoma.

6.
Handb Exp Pharmacol ; 272: 245-265, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34595582

RESUMO

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by peripheral immune cell infiltration into the brain and spinal cord, demyelination, glial cell activation, and neuronal damage. Currently there is no cure for MS, however, available disease-modifying agents minimize inflammation in the CNS by various mechanisms. Approved drugs lessen severity of the disease and delay disease progression, however, they are still suboptimal as patients experience adverse effects and varying efficacies. Additionally, there is only one disease-modifying therapy available for the more debilitating, progressive form of MS. This chapter focuses on the presently-available therapeutics and, importantly, the future directions of MS therapy based on preclinical studies and early clinical trials. Immunosuppression in other neurological disorders including neuromyelitis optica spectrum disorders, myasthenia gravis, and Guillain-Barré syndrome is also discussed.


Assuntos
Esclerose Múltipla , Miastenia Gravis , Neuromielite Óptica , Humanos , Terapia de Imunossupressão , Esclerose Múltipla/tratamento farmacológico , Neuromielite Óptica/tratamento farmacológico , Medula Espinal
7.
Glia ; 69(7): 1767-1781, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33704822

RESUMO

The characterization of the tumor microenvironment (TME) in high grade gliomas (HGG) has generated significant interest in an effort to understand how neoplastic lesions in the central nervous system (CNS) are supported and to devise novel therapeutic targets. The TME of the CNS contains unique and specialized cells, including the resident myeloid cells, microglia. Myeloid involvement in HGG, such as glioblastoma, is associated with poor outcomes. Glioma-associated microglia and infiltrating monocytes/macrophages (GAM) accumulate within the neoplastic lesion where they facilitate tumor growth and drive immunosuppression. However, it has been difficult to differentiate whether microglia and macrophages have similar or distinct roles in pathology, and if the spatial organization of these cells informs outcomes. Here, we characterize the tumor-stroma border and identify peritumoral GAM (PGAM) as a unique subpopulation of GAM. Using data mining and analyses of samples derived from both murine and human sources we show that PGAM exhibit a pro-inflammatory and chemotactic phenotype that is associated with peripheral monocyte recruitment, and decreased overall survival. PGAM act as a unique subset of GAM at the tumor-stroma interface. We define a novel gene signature to identify these cells and suggest that PGAM constitute a cellular target of the TME.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Animais , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Glioma/patologia , Macrófagos/patologia , Camundongos , Microglia/patologia , Microambiente Tumoral
8.
Sci Rep ; 11(1): 5057, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658642

RESUMO

The efficacy of combining radiation therapy with immune checkpoint inhibitor blockade to treat brain tumors is currently the subject of multiple investigations and holds significant therapeutic promise. However, the long-term effects of this combination therapy on the normal brain tissue are unknown. Here, we examined mice that were intracranially implanted with murine glioma cell line and became long-term survivors after treatment with a combination of 10 Gy cranial irradiation (RT) and anti-PD-1 checkpoint blockade (aPD-1). Post-mortem analysis of the cerebral hemisphere contralateral to tumor implantation showed complete abolishment of hippocampal neurogenesis, but neural stem cells were well preserved in subventricular zone. In addition, we observed a drastic reduction in the number of mature oligodendrocytes in the subcortical white matter. Importantly, this observation was evident specifically in the combined (RT + aPD-1) treatment group but not in the single treatment arm of either RT alone or aPD-1 alone. Elimination of microglia with a small molecule inhibitor of colony stimulated factor-1 receptor (PLX5622) prevented the loss of mature oligodendrocytes. These results identify for the first time a unique pattern of normal tissue changes in the brain secondary to combination treatment with radiotherapy and immunotherapy. The results also suggest a role for microglia as key mediators of the adverse treatment effect.


Assuntos
Anticorpos/administração & dosagem , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/radioterapia , Irradiação Craniana/métodos , Glioma/mortalidade , Glioma/radioterapia , Inibidores de Checkpoint Imunológico/administração & dosagem , Imunoterapia/métodos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Terapia Combinada/métodos , Modelos Animais de Doenças , Glioma/metabolismo , Glioma/patologia , Hospedeiro Imunocomprometido , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Compostos Orgânicos/administração & dosagem , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Distribuição Aleatória , Taxa de Sobrevida , Resultado do Tratamento
9.
Int J Mol Sci ; 21(22)2020 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-33187183

RESUMO

Despite significant strides made in understanding the pathophysiology of high-grade gliomas over the past two decades, most patients succumb to these neoplasias within two years of diagnosis. Furthermore, there are various co-morbidities associated with glioma and standard of care treatments. Emerging evidence suggests that aberrant glutamate secretion in the glioma microenvironment promotes tumor progression and contributes to the development of co-morbidities, such as cognitive defects, epilepsy, and widespread neurodegeneration. Recent data clearly illustrate that neurons directly synapse onto glioma cells and drive their proliferation and spread via glutamatergic action. Microglia are central nervous system-resident myeloid cells, modulate glioma growth, and possess the capacity to prune synapses and encourage synapse formation. However, current literature has yet to investigate the potential role of microglia in shaping synapse formation between neurons and glioma cells. Herein, we present the literature concerning glutamate's role in glioma progression, involving hyperexcitability and excitotoxic cell death of peritumoral neurons and stimulation of glioma proliferation and invasion. Furthermore, we discuss instances in which microglia are more likely to sculpt or encourage synapse formation during glioma treatment and propose studies to delineate the role of microglia in synapse formation between neurons and glioma cells. The sex-dependent oncogenic or oncolytic actions of microglia and myeloid cells, in general, are considered in addition to the functional differences between microglia and macrophages in tumor progression. We also put forth tractable methods to safely perturb aberrant glutamatergic action in the tumor microenvironment without significantly increasing the toxicities of the standard of care therapies for glioma therapy.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Microglia/patologia , Neurônios/patologia , Microambiente Tumoral/fisiologia , Animais , Carcinogênese/patologia , Progressão da Doença , Humanos , Macrófagos/patologia
10.
Sci Rep ; 10(1): 19333, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33168944

RESUMO

Multiple sclerosis (MS) is an autoimmune disease characterized by infiltration of peripheral immune cells into the central nervous system, demyelination, and neuronal damage. There is no cure for MS, but available disease-modifying therapies can lessen severity and delay progression. However, current therapies are suboptimal due to adverse effects. Here, we investigate how the FDA-approved antihypertensive drug, guanabenz, which has a favorable safety profile and was recently reported to enhance oligodendrocyte survival, exerts effects on immune cells, specifically microglia and macrophages. We first employed the experimental autoimmune encephalomyelitis (EAE) model and observed pronounced immunomodulation evident by a reduction in pro-inflammatory microglia and macrophages. When guanabenz was administered in the cuprizone model, in which demyelination is less dependent upon immune cells, we did not observe improvements in remyelination, oligodendrocyte numbers, and effects on microglial activation were less dramatic. Thus, guanabenz may be a promising therapeutic to minimize inflammation without exerting severe off-target effects.


Assuntos
Doenças Desmielinizantes , Guanabenzo/farmacologia , Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Animais , Anti-Hipertensivos/farmacologia , Antígeno B7-2/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Encefalomielite Autoimune Experimental/imunologia , Feminino , Inflamação/tratamento farmacológico , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Oligodendroglia/efeitos dos fármacos , Segurança do Paciente , Fagocitose , Remielinização/efeitos dos fármacos
11.
J Neurooncol ; 147(3): 547-555, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32215786

RESUMO

INTRODUCTION: Glioblastoma multiforme (GBM) is a deadly brain tumor with a short expected median survival, despite current standard-of-care treatment. We explored the combination of intermediate stereotactic dose radiation therapy and immune checkpoint inhibitor therapy as a novel treatment strategy for GBM. METHODS: Glioma xenograft-bearing mice were exposed to high dose brain-directed radiation (10 Gy single exposure) as well as mouse anti-PD-1 antibody. The tumor-bearing animals were randomized to four groups: no treatment, radiation alone, anti-PD-1 alone, and radiation + anti-PD-1. Survival was followed, and tumor growth was monitored using MRI. Immunohistochemistry, gene expression arrays, and flow cytometry were used to characterize the treatment-induced effects. Pharmacologic inhibitors of T-lymphocytes, bone marrow derived macrophages, and microglia were used to assess the respective roles of different immune populations in observed treatment effects. RESULTS: We found the combined treatment with high dose radiation and immunotherapy to be highly effective with a 75% complete pathologic response and dramatically improved survival outcomes. We found both CD8+ T-cells and macrophages to be necessary for the full effect of combined therapy, with T lymphocytes appearing to play a role early on and macrophages mediating a later phase of the combined treatment effect. Radiation treatment appeared to trigger macrophage repolarization, increasing M1/M2 ratio. CONCLUSIONS: These findings point to a novel immunologic mechanism underlying the interaction between radiotherapy and immunotherapy. They also provide the basis for clinical investigation of immunogenic dose radiation in combination with immune checkpoint blockade as a potential treatment approach for newly diagnosed high grade gliomas.


Assuntos
Neoplasias Encefálicas/radioterapia , Glioma/radioterapia , Inibidores de Checkpoint Imunológico/uso terapêutico , Macrófagos/efeitos dos fármacos , Macrófagos/efeitos da radiação , Radiocirurgia/métodos , Animais , Neoplasias Encefálicas/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Expressão Gênica , Glioma/imunologia , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Doses de Radiação , Análise de Sobrevida
12.
J Appl Toxicol ; 39(10): 1413-1423, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31319435

RESUMO

Lunar regolith samples collected during previous Apollo missions were found to contain components that were established to be toxic to humans; however, the health effects due to inhalation of lunar soil as a whole are still unknown. Macrophages residing in the alveolar sacs of the lungs constitute one of the last lines of defense against inhaled particulates before entry into the bloodstream. Here, we examine the macrophage response to lunar simulants that are similar in chemical composition to the lunar regolith. We assess cytotoxicity, cellular morphology, phagocytosis of simulants and expression of inflammatory markers. Overall, the exposure of macrophages to lunar simulants results in moderate cytotoxicity and marked alteration of cell morphology and uptake of the simulants. Interestingly, simulant exposure decreased proinflammatory gene expression, but may induce an anti-inflammatory phenotype in the cells. These results illustrate that although macrophages phagocytose lunar simulants as a protective response, the simulants do induce a degree of macrophage cell death. Our study reveals some toxicity associated with lunar simulants and supports further evaluation of the inhalation of lunar regolith to understand the risks of exposure fully.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Poeira Cósmica/efeitos adversos , Macrófagos/efeitos dos fármacos , Lua , Solo/química , Voo Espacial , Administração por Inalação , Humanos
13.
Opt Express ; 27(13): 17620-17637, 2019 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-31252719

RESUMO

Single-photon-excitation-based miniaturized microscope, or miniscope, has recently emerged as a powerful tool for imaging neural ensemble activities in freely moving animals. In the meanwhile, this highly flexible and implantable technology promises great potential for studying a broad range of cells, tissues and organs. To date, however, applications have been largely limited by the properties of the imaging modality. It is therefore highly desirable for a method generally applicable for processing miniscopy images, enabling and extending the applications to diverse anatomical and functional traits, spanning various cell types in the brain and other organs. We report an image processing approach, termed BSSE, for background suppression and signal enhancement for miniscope image processing. The BSSE method provides a simple, automatic solution to the intrinsic challenges of overlapping signals, high background and artifacts in miniscopy images. We validated the method by imaging synthetic structures and various biological samples of brain, tumor, and kidney tissues. The work represents a generally applicable tool for miniscopy technology, suggesting broader applications of the miniaturized, implantable and flexible technology for biomedical research.

14.
F1000Res ; 82019.
Artigo em Inglês | MEDLINE | ID: mdl-30863536

RESUMO

Multiple sclerosis (MS) is a major cause of disability in young adults. Following an unknown trigger (or triggers), the immune system attacks the myelin sheath surrounding axons, leading to progressive nerve cell death. Antibodies and small-molecule drugs directed against B cells have demonstrated good efficacy in slowing progression of the disease. This review focusses on small-molecule drugs that can affect B-cell biology and may have utility in disease management. The risk genes for MS are examined from the drug target perspective. Existing small-molecule therapies for MS with B-cell actions together with new drugs in development are described. The potential for experimental molecules with B-cell effects is also considered. Small molecules can have diverse actions on B cells and be cytotoxic, anti-inflammatory and anti-viral. The current B cell-directed therapies often kill B-cell subsets, which can be effective but lead to side effects and toxicity. A deeper understanding of B-cell biology and the effect on MS disease should lead to new drugs with better selectivity, efficacy, and an improved safety profile. Small-molecule drugs, once the patent term has expired, provide a uniquely sustainable form of healthcare.


Assuntos
Antineoplásicos , Linfócitos B , Esclerose Múltipla , Animais , Linfócitos B/fisiologia , Camundongos , Esclerose Múltipla/imunologia , Esclerose Múltipla/terapia , Bibliotecas de Moléculas Pequenas
15.
Oncotarget ; 9(86): 35655-35665, 2018 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-30479695

RESUMO

High grade gliomas, including glioblastoma (GB), are devastating malignancies with very poor prognosis. Over the course of the last decade, there has been a failure to develop new treatments for GB. Reasons for this failure include the lack of validation of novel molecular targets, which are often characterized in animal models and directly transposed to human trials. Here we build on our previous findings, which describe how the multi-functional co-receptor Neuropilin-1 (NRP1) signals through glioma associated microglia/macrophages (GAMS) to promote murine glioma, and investigate NRP1 expression in human glioma. Clinical and gene expression data were obtained via The Cancer Genome Atlas (TCGA), and analyzed using R statistical software. Additionally, CIBERSORT in silico deconvolution was used to determine fractions of immune cell sub-populations within the gene expression datasets. We find that NRP1 expression is correlated with poor prognosis, glioma grade, and associates with the mesenchymal GB subtype. In human GB, NRP1 expression is highly correlated with markers of monocytes/macrophages, as well as genes that contribute to the pro-tumorigenic phenotype of these cells.

16.
Spinal Cord ; 56(8): 733-740, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29904189

RESUMO

STUDY DESIGN: Experimental study. OBJECTIVES: To evaluate the efficacy of Angiotensin-converting enzyme inhibitor Ramipril, as a mitigator of radiation-induced spinal cord injury. SETTING: Stony Brook University, Stony Brook, NY, USA. METHODS: Total of 22 rats were irradiated with single doses of 23.6-33 Gy at the C4-T2 spinal levels. After irradiation, the rats were randomized to the radiation only control group and the Ramipril-treated (radiation + Ramipril) experimental group. Ramipril 1.5 mg/kg/day was given in the drinking water starting 1 week after radiation through the study duration. RESULTS: All the rats irradiated with 28.5-33 Gy became paralyzed at 125 ± 4 days, whereas no rats became paralyzed after 23.6 Gy. The time to develop paralysis was delayed to 135 ± 4 days in Ramipril-treated group (P < 0.001). H&E and LFB showed microscopic structural restoration and remyelination with Ramipril treatment. VEGF expression was increased in the irradiated spinal cord, and the number of VEGF-positive cells was significantly decreased by Ramipril treatment (P < 0.001). Immunohistochemical stain with Iba-1 showed increased microglial infiltration in the irradiated spinal cords. The number of Iba-1-positive microglia was significantly reduced by Ramipril treatment (P < 0.05). CONCLUSION: Ramipril reduced the rate of paralysis even at the paralysis-inducing radiation doses. It also significantly delayed the onset of paralysis. Neuroinflammation and endothelial cell damage may be the key mediators of radiation injury. Ramipril can be readily translatable to clinical application as a mitigatory of radiotherapeutic toxicity.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Microglia/efeitos dos fármacos , Lesões Experimentais por Radiação/tratamento farmacológico , Ramipril/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/etiologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Proteínas dos Microfilamentos/metabolismo , Microglia/patologia , Microglia/fisiologia , Microglia/efeitos da radiação , Paralisia/tratamento farmacológico , Paralisia/etiologia , Paralisia/patologia , Paralisia/fisiopatologia , Lesões Experimentais por Radiação/patologia , Lesões Experimentais por Radiação/fisiopatologia , Distribuição Aleatória , Ratos Endogâmicos F344 , Remielinização/efeitos dos fármacos , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Regeneração da Medula Espinal/efeitos dos fármacos
17.
J Med Chem ; 61(9): 4135-4154, 2018 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-29648813

RESUMO

We report the design, synthesis, and biological evaluation of some potent small-molecule neuropilin-1 (NRP1) antagonists. NRP1 is implicated in the immune response to tumors, particularly in Treg cell fragility, required for PD1 checkpoint blockade. The design of these compounds was based on a previously identified compound EG00229. The design of these molecules was informed and supported by X-ray crystal structures. Compound 1 (EG01377) was identified as having properties suitable for further investigation. Compound 1 was then tested in several in vitro assays and was shown to have antiangiogenic, antimigratory, and antitumor effects. Remarkably, 1 was shown to be selective for NRP1 over the closely related protein NRP2. In purified Nrp1+, FoxP3+, and CD25+ populations of Tregs from mice, 1 was able to block a glioma-conditioned medium-induced increase in TGFß production. This comprehensive characterization of a small-molecule NRP1 antagonist provides the basis for future in vivo studies.


Assuntos
Imunomodulação/efeitos dos fármacos , Neuropilina-1/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Camundongos , Modelos Moleculares , Conformação Molecular , Ácidos Pentanoicos/química , Ácidos Pentanoicos/farmacologia , Bibliotecas de Moléculas Pequenas/química , Linfócitos T Reguladores/imunologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
18.
Proc Natl Acad Sci U S A ; 115(13): 3482-3487, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29531087

RESUMO

Endocannabinoids (eCBs) are lipid-signaling molecules involved in the regulation of numerous behaviors and physiological functions. Released by postsynaptic neurons, eCBs mediate retrograde modulation of synaptic transmission and plasticity by activating presynaptic cannabinoid receptors. While the cellular mechanisms by which eCBs control synaptic function have been well characterized, the mechanisms controlling their retrograde synaptic transport remain unknown. Here, we demonstrate that fatty-acid-binding protein 5 (FABP5), a canonical intracellular carrier of eCBs, is indispensable for retrograde eCB transport in the dorsal raphe nucleus (DRn). Thus, pharmacological inhibition or genetic deletion of FABP5 abolishes both phasic and tonic eCB-mediated control of excitatory synaptic transmission in the DRn. The blockade of retrograde eCB signaling induced by FABP5 inhibition is not mediated by impaired cannabinoid receptor function or reduced eCB synthesis. These findings indicate that FABP5 is essential for retrograde eCB signaling and may serve as a synaptic carrier of eCBs at central synapses.


Assuntos
Ácidos Araquidônicos/metabolismo , Endocanabinoides/farmacologia , Proteínas de Ligação a Ácido Graxo/fisiologia , Ácido Glutâmico/metabolismo , Glicerídeos/metabolismo , Proteínas de Neoplasias/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Células Cultivadas , Endocanabinoides/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/metabolismo , Sinapses/efeitos dos fármacos
19.
AIMS Allergy Immunol ; 2(1): 24-44, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-32914058

RESUMO

Immunotherapies are becoming a promising strategy for malignant disease. Selectively directing host immune responses to target cancerous tissue is a milestone of human health care. The roles of the innate and adaptive immune systems in both cancer progression and elimination are now being realized. Defining the immune cell environment and identifying the contributions of each sub-population of these cells has lead to an understanding of the immunotherapeutic processes, and demonstrated the potential of the immune system to drive cancer shrinkage and sustained immunity against disease. Poorly treated diseases, such as high-grade glioma, suffer from lack of therapeutic efficacy and rapid progression. Immunotherapeutic success in other solid malignancies, such as melanoma, now provides the principals for which this treatment paradigm can be adapted for primary brain cancers. The central nervous system is complex, and relative contributions of immune sub-populations to high grade glioma progression are not fully characterized. Here, we summarize recent research in both animal and humans which add to the knowledge base of how innate and adaptive immune cells contribute to glioma progression, and outline work which has demonstrated their potential to elicit anti-tumorigenic responses. Additionally, we highlight Neuropilin 1, a cell surface receptor protein, describe its signaling functions in the context of immunity, and point to its potential to slow glioma progression.

20.
J Neurol ; 265(4): 741-756, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29209782

RESUMO

Gliomas are primary malignancies of the brain. Tumors are staged based on malignancy, nuclear atypia, and infiltration of the surrounding brain parenchyma. Tumors are often diagnosed once patients become symptomatic, at which time the lesion is sizable. Glioblastoma (grade IV glioma) is highly aggressive and difficult to treat. Most tumors are diagnosed de novo. The gold standard of therapy, implemented over a decade ago, consists of fractionated radiotherapy and temozolomide, but unfortunately, chemotherapeutic resistance arises. Recurrence is common after initial therapy. The tumor microenvironment plays a large role in cancer progression and its manipulation can repress progression. The advent and implementation of immunotherapy, via manipulation and activation of cytotoxic T cells, have had an outstanding impact on reducing morbidity and mortality associated with peripheral cancers under certain clinical circumstances. An arsenal of immunotherapeutics is currently under clinical investigation for safety and efficacy in the treatment of newly diagnosed and recurrent high grade gliomas. These immunotherapeutics encompass antibody-drug conjugates, autologous infusions of modified chimeric antigen receptor expressing T cells, peptide vaccines, autologous dendritic cell vaccines, immunostimulatory viruses, oncolytic viruses, checkpoint blockade inhibitors, and drugs which alter the behavior of innate immune cells. Effort is focusing on determining which patient populations will benefit the most from these treatments and why. Research addressing synergism between treatment options is gaining attention. While advances in the treatment of glioma stagnated in the past, we may see a considerable evolution in the management of the disease in the upcoming years.


Assuntos
Pesquisa Biomédica , Neoplasias Encefálicas/terapia , Glioma/terapia , Imunoterapia/métodos , Neoplasias Encefálicas/imunologia , Terapia Combinada , Glioma/imunologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA