Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 7(4)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35015729

RESUMO

Monocyte-derived macrophages (MDMs) are key players in tissue homeostasis and diseases regulated by a variety of signaling molecules. Recent literature has highlighted the ability for biogenic amines to regulate macrophage functions, but the mechanisms governing biogenic amine signaling in and around immune cells remain nebulous. In the CNS, biogenic amine transporters are regarded as the master regulators of neurotransmitter signaling. While we and others have shown that macrophages express these transporters, relatively little is known of their function in these cells. To address these knowledge gaps, we investigated the function of norepinephrine transporter (NET) and dopamine transporter (DAT) on human MDMs. We found that both NET and DAT are present and can uptake substrate from the extracellular space at baseline. Not only was DAT expressed in cultured MDMs, but it was also detected in a subset of intestinal macrophages in situ. Surprisingly, we discovered a NET-independent, DAT-mediated immunomodulatory mechanism in response to LPS. LPS induced reverse transport of dopamine through DAT, engaging an autocrine/paracrine signaling loop that regulated the macrophage response. Removing this signaling loop enhanced the proinflammatory response to LPS. Our data introduce a potential role for DAT in the regulation of innate immunity.


Assuntos
Aminas Biogênicas/metabolismo , Transporte Biológico/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Regulação da Expressão Gênica , Macrófagos/metabolismo , RNA/genética , Adulto , Idoso , Proteínas da Membrana Plasmática de Transporte de Dopamina/biossíntese , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Feminino , Humanos , Macrófagos/patologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
2.
NPJ Parkinsons Dis ; 7(1): 62, 2021 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-34285243

RESUMO

Most, if not all, peripheral immune cells in humans and animals express tyrosine hydroxylase (TH), the rate limiting enzyme in catecholamine synthesis. Since TH is typically studied in the context of brain catecholamine signaling, little is known about changes in TH production and function in peripheral immune cells. This knowledge gap is due, in part, to the lack of an adequately sensitive assay to measure TH in immune cells expressing lower TH levels compared to other TH expressing cells. Here, we report the development of a highly sensitive and reproducible Bio-ELISA to quantify picogram levels of TH in multiple model systems. We have applied this assay to monocytes isolated from blood of persons with Parkinson's disease (PD) and to age-matched, healthy controls. Our study unexpectedly revealed that PD patients' monocytes express significantly higher levels of TH protein in peripheral monocytes relative to healthy controls. Tumor necrosis factor (TNFα), a pro-inflammatory cytokine, has also been shown to be increased in the brains and peripheral circulation in human PD, as well as in animal models of PD. Therefore, we investigated a possible connection between higher levels of TH protein and the known increase in circulating TNFα in PD. Monocytes isolated from healthy donors were treated with TNFα or with TNFα in the presence of an inhibitor. Tissue plasminogen activator (TPA) was used as a positive control. We observed that TNFα stimulation increased both the number of TH+ monocytes and the quantity of TH per monocyte, without increasing the total numbers of monocytes. These results revealed that TNFα could potentially modify monocytic TH production and serve a regulatory role in peripheral immune function. The development and application of a highly sensitive assay to quantify TH in both human and animal cells will provide a novel tool for further investigating possible PD immune regulatory pathways between brain and periphery.

3.
J Pharmacol Exp Ther ; 375(1): 154-160, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32238454

RESUMO

Mononuclear macrophages derived from the bone marrow (myeloid cells) are key cellular components of the innate immune system in different organs. In this minireview, we are focused on both brain and blood macrophages, known as microglia and monocytes, respectively. We provide a succinct summary of the cells' functions under both normal and pathologic conditions, with particular reference to common neurodegenerative disorders, such as Alzheimer and Parkinson disease. SIGNIFICANCE STATEMENT: In this minireview, we aim to summarize available literature on microglial and myeloid involvement in CNS disease, directing the reader toward relevant and translatable interpretations of myeloid cell function in CNS health and neurodegeneration.


Assuntos
Doença de Alzheimer/patologia , Sistema Nervoso Central/imunologia , Microglia/patologia , Células Mieloides/patologia , Neuroimunomodulação , Doença de Parkinson/patologia , Doença de Alzheimer/sangue , Doença de Alzheimer/imunologia , Animais , Sistema Nervoso Central/patologia , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Microglia/imunologia , Monócitos/imunologia , Monócitos/patologia , Células Mieloides/imunologia , Doença de Parkinson/sangue , Doença de Parkinson/imunologia
4.
Biomaterials ; 188: 144-159, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343257

RESUMO

The use of intracortical microelectrode arrays has gained significant attention in being able to help restore function in paralysis patients and study the brain in various neurological disorders. Electrode implantation in the cortex causes vasculature or blood-brain barrier (BBB) disruption and thus elicits a foreign body response (FBR) that results in chronic inflammation and may lead to poor electrode performance. In this study, a comprehensive insight into the acute molecular mechanisms occurring at the Utah electrode array-tissue interface is provided to understand the oxidative stress, neuroinflammation, and neurovascular unit (astrocytes, pericytes, and endothelial cells) disruption that occurs following microelectrode implantation. Quantitative real time polymerase chain reaction (qRT-PCR) was used to quantify the gene expression at acute time-points of 48-hr, 72-hr, and 7-days for factors mediating oxidative stress, inflammation, and BBB disruption in rats implanted with a non-functional 4 × 4 Utah array in the somatosensory cortex. During vascular disruption, free iron released into the brain parenchyma can exacerbate the FBR, leading to oxidative stress and thus further contributing to BBB degradation. To reduce the free iron released into the brain tissue, the effects of an iron chelator, deferoxamine mesylate (DFX), was also evaluated.


Assuntos
Barreira Hematoencefálica/patologia , Desferroxamina/uso terapêutico , Eletrodos Implantados/efeitos adversos , Corpos Estranhos/tratamento farmacológico , Corpos Estranhos/etiologia , Quelantes de Ferro/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Corpos Estranhos/metabolismo , Corpos Estranhos/patologia , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley
5.
Curr Opin Immunol ; 29: 93-6, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24908174

RESUMO

Microglial cells comprising the brain's immune system are essential for ensuring neuroprotection in the normal and pathological CNS. On the basis of histopathological observations in human brain, we believe that the ability of microglia to provide neuroprotection deteriorates as our brains get older and that such CNS immune senescence is a major factor contributing to the development of aging-related neurodegenerative diseases, notably Alzheimer's disease. The idea is consistent with the fact that immune senescence occurs naturally in the periphery, rendering the elderly people more susceptible to infections and cancers. There is an analogous situation in the brain, except that here the main impact comes down to diminished neuroprotection and resultant neurodegeneration.


Assuntos
Doenças Neurodegenerativas/imunologia , Envelhecimento , Animais , Senescência Celular , Humanos , Inflamação/imunologia
6.
J Neural Eng ; 9(5): 056015, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23010756

RESUMO

For nearly 55 years, tungsten microwires have been widely used in neurophysiological experiments in animal models to chronically record neuronal activity. While tungsten microwires initially provide stable recordings, their inability to reliably record high-quality neural signals for tens of years has limited their efficacy for neuroprosthetic applications in humans. Comprehensive understanding of the mechanisms of electrode performance and failure is necessary for developing next generation neural interfaces for humans. In this study, we evaluated the abiotic (electrophysiology, impedance, electrode morphology) and biotic (microglial reactivity, blood-brain barrier disruption, biochemical markers of axonal injury) effects of 16-channel, 50 µm diameter, polyimide insulated tungsten microwires array for implant durations that ranged from acute to up to 9 months in 25 rats. Daily electrode impedance spectroscopy, electrophysiological recordings, blood and cerebrospinal fluid (CSF) withdrawals, and histopathological analysis were performed to study the time-varying effects of chronic electrode implantation. Structural changes at the electrode recording site were observed as early as within 2-3 h of electrode insertion. Abiotic analysis indicated the first 2-3 weeks following surgery was the most dynamic period in the chronic electrode lifetime as there were greater variations in the electrode impedance, functional electrode performance, and the structural changes occurring at the electrode recording tips. Electrode recording site deterioration continued for the long-term chronic animals as insulation damage occurred and recording surface became more recessed over time. In general, electrode impedance and functional performance had smaller daily variations combined with reduced electrode recording site changes during the chronic phase. Histopathological studies were focused largely on characterizing microglial cell responses to electrode implantation. We found that activated microglia were present near the electrode tracks in all non-acute animals studied, thus indicating presence of a neuroinflammatory response regardless of post-implantation survival times and electrode performance. Conversely, dystrophic microglia detectable as fragmented cells were found almost exclusively in acute animals surviving only few hours after implantation. While there was no consistent relationship between microglial cell responses and electrode performance, we noticed co-occurrence of high ferritin expression, intraparenchymal bleeding, and microglial degeneration suggesting presence of excessive oxidative stress via Fenton chemistry. Biochemical analysis indicated that these electrodes always caused a persistent release of axonal injury biomarkers even several months after implantation suggesting persistent tissue damage. Our study suggests that mechanisms of electrode failure are multi-factorial involving both abiotic and biotic parameters. Since these failure modes occur concurrently and cannot be isolated from one another, the lack of consistent relationship between electrode performance and microglial responses in our results suggest that one or more of the abiotic factors were equally responsible for degradation in electrode performance over long periods of time.


Assuntos
Córtex Cerebral/fisiologia , Córtex Cerebral/ultraestrutura , Eletrodos Implantados/normas , Falha de Equipamento , Microeletrodos/normas , Tungstênio , Animais , Impedância Elétrica , Masculino , Ratos , Ratos Sprague-Dawley , Tungstênio/química
7.
PLoS One ; 7(4): e34097, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496779

RESUMO

Transplantation of neural stems cells (NSCs) could be a useful means to deliver biologic therapeutics for late-stage Alzheimer's disease (AD). In this study, we conducted a small preclinical investigation of whether NSCs could be modified to express metalloproteinase 9 (MMP9), a secreted protease reported to degrade aggregated Aß peptides that are the major constituents of the senile plaques. Our findings illuminated three issues with using NSCs as delivery vehicles for this particular application. First, transplanted NSCs generally failed to migrate to amyloid plaques, instead tending to colonize white matter tracts. Second, the final destination of these cells was highly influenced by how they were delivered. We found that our injection methods led to cells largely distributing to white matter tracts, which are anisotropic conduits for fluids that facilitate rapid distribution within the CNS. Third, with regard to MMP9 as a therapeutic to remove senile plaques, we observed high concentrations of endogenous metalloproteinases around amyloid plaques in the mouse models used for these preclinical tests with no evidence that the NSC-delivered enzymes elevated these activities or had any impact. Interestingly, MMP9-expressing NSCs formed substantially larger grafts. Overall, we observed long-term survival of NSCs in the brains of mice with high amyloid burden. Therefore, we conclude that such cells may have potential in therapeutic applications in AD but improved targeting of these cells to disease-specific lesions may be required to enhance efficacy.


Assuntos
Amiloidose/prevenção & controle , Encéfalo/patologia , Modelos Animais de Doenças , Metaloproteinase 9 da Matriz/metabolismo , Fibras Nervosas Mielinizadas/patologia , Células-Tronco Neurais/transplante , Placa Amiloide/prevenção & controle , Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/enzimologia , Amiloidose/patologia , Animais , Encéfalo/metabolismo , Células Cultivadas , Técnicas Imunoenzimáticas , Lentivirus/genética , Camundongos , Fibras Nervosas Mielinizadas/metabolismo , Células-Tronco Neurais/citologia , Placa Amiloide/enzimologia , Placa Amiloide/patologia
8.
Neurobiol Aging ; 33(1): 195.e1-12, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20580465

RESUMO

To understand how microglial cell function may change with aging, various protocols have been developed to isolate microglia from the young and aged central nervous system (CNS). Here we report modification of an existing protocol that is marked by less debris contamination and improved yields and demonstrate that microglial functions are varied and dependent on age. Specifically, we found that microglia from aged mice constitutively secrete greater amounts of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) relative to microglia from younger mice and are less responsive to stimulation. Also, microglia from aged mice have reduced glutathione levels and internalize less amyloid beta peptide (Aß) while microglia from mice of all ages do not retain the amyloid beta peptide for a significant length of time. These studies offer further support for the idea that microglial cell function changes with aging. They suggest that microglial Aß phagocytosis results in Aß redistribution rather than biophysical degradation in vivo and thereby provide mechanistic insight to the lack of amyloid burden elimination by parenchymal microglia in aged adults and those suffering from Alzheimer's disease.


Assuntos
Envelhecimento/fisiologia , Encéfalo/citologia , Microglia/fisiologia , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/fisiologia , Sobrevivência Celular , Células Cultivadas , Glutationa/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Fagocitose , Fator de Necrose Tumoral alfa/metabolismo
9.
Brain Res ; 1319: 131-41, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20059990

RESUMO

The neuronal pathology caused by neonatal infection of rats with the PVC-211 murine leukemia virus (PVC-211 MuLV) and its underlying mechanisms are not well defined even though a loss of neurons and spongiform neurodegeneration has been reported to accompany the disease. Here we sought to identify sites of neurodegeneration using microglial reactivity as an indirect marker and to characterize microglial activation during disease progression. Using a panel of microglial antibodies including Iba1, OX-42, ED1, and anti-ferritin, we have studied the response of microglial cells to neonatal CNS infection with PVC-211 at post-infection survival times 7, 14, 21, and 28 days. We found that microglial activation occurred primarily in the spinal cord and brainstem where it gradually increased in intensity over the time course of this study. Other brain areas were relatively unremarkable in their microglial reaction to viral infection within this time frame. However, the presence of activated microglial cells was not correlated directly with the presence of viral glycoprotein (gp70), which was expressed in endothelial cells throughout the CNS. Although double-labeling of microglia with Iba1 and ED1 revealed numerous actively phagocytic microglia during disease progression, not all activated microglia were ED1-positive. In addition to the intense microglial activation, we found increased ferritin expression sporadically throughout the virus-infected brain. The ferritin-positive cells were mostly microglia that exhibited dystrophic changes and likely represented a degenerating subpopulation of microglial cells. Thus, activated microglia can co-exist with degenerating microglia in the same brain region. We attempted to localize degenerating neurons or neurites using Fluoro-Jade, anti-tau, and anti-alpha synuclein staining, but none of these procedures yielded results to indicate obvious neuronal pathology. We conclude that the visualization of microglial activation is a more sensitive measure of neuronal perturbations than direct detection of neuronal pathology which may be subtle and not produce overt degenerative changes.


Assuntos
Infecções do Sistema Nervoso Central/fisiopatologia , Vírus da Leucemia Murina , Microglia/fisiologia , Degeneração Neural/fisiopatologia , Infecções por Retroviridae/fisiopatologia , Infecções Tumorais por Vírus/fisiopatologia , Animais , Animais Recém-Nascidos , Encéfalo/fisiopatologia , Progressão da Doença , Ferritinas/metabolismo , Neuritos/fisiologia , Neurônios/fisiologia , Ratos , Ratos Endogâmicos F344 , Medula Espinal/fisiopatologia , Fatores de Tempo
10.
Acta Neuropathol ; 119(1): 89-105, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20012873

RESUMO

The past 20 years have seen a gain in knowledge on microglia biology and microglia functions in disease that exceeds the expectations formulated when the microglia "immune network" was introduced. More than 10,000 articles have been published during this time. Important new research avenues of clinical importance have opened up such as the role of microglia in pain and in brain tumors. New controversies have also emerged such as the question of whether microglia are active or reactive players in neurodegenerative disease conditions, or whether they may be victims themselves. Premature commercial interests may be responsible for some of the confusion that currently surrounds microglia in both the Alzheimer and Parkinson's disease research fields. A critical review of the literature shows that the concept of "(micro)glial inflammation" is still open to interpretation, despite a prevailing slant towards a negative meaning. Perhaps the most exciting foreseeable development concerns research on the role of microglia in synaptic plasticity, which is expected to yield an answer to the question whether microglia are the brain's electricians. This review provides an analysis of the latest developments in the microglia field.


Assuntos
Microglia/patologia , Microglia/fisiologia , Animais , Sistema Nervoso Central/patologia , Sistema Nervoso Central/fisiologia , Sistema Nervoso Central/fisiopatologia , Doenças do Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/fisiopatologia , Humanos , Modelos Neurológicos
11.
Neoplasia ; 10(8): 804-16, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18680882

RESUMO

The thymidine analog bromodeoxyuridine (BrdU) is incorporated into newly synthesized DNA and has been shown to increase the susceptibility of incorporating cells to ionizing radiation. However, in the absence of secondary stressors, BrdU is thought to substitute relatively benignly for thymidine and is commonly used to "birth-date" proliferative cells. We report a novel antiproliferative effect of BrdU on cancer cells, which is independent of its role in radiosensitization. A single, brief in vitro exposure to BrdU induces a profound and sustained reduction in the proliferation rate of all cancer cells examined. Cells do not die but variably up-regulate some senescence-associated proteins as they accumulate in the G1 phase of the cell cycle. Bromodeoxyuridine also impairs the proliferative capacity of primary tumor-initiating human glioma cells and may therefore represent a means of targeting cancer stem cells. Finally, conservative in vivo BrdU regimens--in the absence of any other treatment--significantly suppress the progression of gliomas in the highly aggressive, syngeneic RG2 model. These results suggest that BrdU may have an important role as an adjunctive therapeutic for a wide variety of cancers based on new insights into its effect as a negative regulator of cell cycle progression.


Assuntos
Bromodesoxiuridina/farmacologia , Glioma/tratamento farmacológico , Neoplasias Experimentais/tratamento farmacológico , Administração Oral , Animais , Bromodesoxiuridina/administração & dosagem , Bromodesoxiuridina/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Glioma/patologia , Humanos , Injeções Intraperitoneais , Injeções Subcutâneas , Masculino , Neoplasias Experimentais/patologia , Nucleosídeos/farmacologia , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Neuroimmunol ; 198(1-2): 98-105, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18508133

RESUMO

Human glioblastoma multiforme (GBM) is the most malignant form of human brain tumors. A characteristic of GBM is the marked presence of tumor infiltrated microglia/macrophages and lymphocytes. The goal of this study was directed toward understanding the role of the chemokine system CX3CL1 and its receptor CX3CR1 in the GL261 murine model of malignant glioma. In situ hybridization analysis identified CX3CL1 and CX3CR1 expression in GL261 tumors. The impact of CX3CR1 deletion on the growth of intracranial GL261 gliomas and associated immune cell infiltration was evaluated in CX3CR1 gene-disrupted C57BL/6 mice. A slight increase in the tumor growth rate in CX3CR1-/- mice was evident with similar numbers of microglia and CD4+, CD8+, FoxP3+, or Ly49G2+ lymphocytes within tumors established in CX3CR1 +/- and -/- mice. These data indicate that CX3CR1 has little or no effects on either gliomagenesis or the migration of microglia and lymphocytes into GL261 tumors.


Assuntos
Quimiocina CX3CL1/metabolismo , Glioma/metabolismo , Glioma/patologia , Linfócitos/imunologia , Microglia/imunologia , Receptores de Quimiocinas/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Receptor 1 de Quimiocina CX3C , Movimento Celular , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Lectinas Tipo C/metabolismo , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Receptores de Quimiocinas/deficiência , Receptores Semelhantes a Lectina de Células NK , Análise de Sobrevida
13.
J Neurooncol ; 85(3): 231-40, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17568998

RESUMO

PURPOSE: Nonspecific stimulation of cells of the immune system may be useful in generating an anti-tumor response for a variety of cancers and may work synergistically with currently available cytotoxic therapies. In this study we examined the response of syngeneic rat gliomas to treatment with several nonspecific stimulators of dendritic cells and macrophages alone or in combination with radiation therapy. EXPERIMENTAL DESIGN: RG-2 gliomas were implanted subcutaneously and treated with intratumoral (IT) injections of the toll-like receptor (TLR) ligands lipopolysaccharide (LPS) and zymosan A (ZymA) and the cytokine granulocyte-macrophage colony stimulating factor (GM-CSF). Combination treatment with IT LPS and single-fraction external beam radiotherapy (EBRT) was also evaluated. RESULTS: Treatment with IT LPS and ZymA delayed tumor growth compared to saline controls. Multiple doses of both substances were superior to single doses, and led to complete tumor regression in 71% (LPS) and 50% (ZymA) of animals. GM-CSF showed no anti-tumor effects in this study. Combinations of IT LPS and EBRT appeared to have a synergistic effect in delaying tumor growth. Rechallenge studies and IT LPS treatment of RG-2 tumors in nude rats suggested the importance of T cells in this treatment paradigm. CONCLUSIONS: Direct IT treatment with the TLR ligands LPS and ZymA are effective in generating an anti-tumor response. These treatments may synergize with cytotoxic therapies such as EBRT, and appear to require T cells for a successful outcome.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Imunoterapia/métodos , Lipopolissacarídeos/administração & dosagem , Zimosan/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/radioterapia , Terapia Combinada , Células Dendríticas/imunologia , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Glioma/imunologia , Glioma/mortalidade , Glioma/radioterapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Injeções Intralesionais , Injeções Subcutâneas , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Masculino , Neoplasias Experimentais , Ratos , Ratos Endogâmicos F344 , Ratos Nus , Tela Subcutânea/patologia , Análise de Sobrevida , Resultado do Tratamento , Zimosan/imunologia
14.
J Neurooncol ; 79(3): 243-53, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16612573

RESUMO

Immunotherapy holds great promise for the treatment of invasive brain tumors, and we are interested specifically in evaluating immune stimulation of microglial cells as one potential strategy. In order to better understand the tumor fighting capabilities of microglial cells, we have compared the responses of syngeneic (Fisher 344) and allogeneic (Wistar) rat strains after intracranial implantation of RG-2 gliomas. Animals were evaluated by clinical examination, magnetic resonance imaging (MRI) and immunohistochemistry for microglial and other immune cell antigens. While lethal RG-2 gliomas developed in all of the Fisher 344 rats, tumors grew variably in the Wistar strain, sometimes reaching considerable sizes, but eventually all of them regressed. Tumor regression was associated with greater numbers of T cells and CD8 positive cells and increases in MHC I and CD4 positive microglia. Our findings suggest that the combined mobilization of peripheral and CNS endogenous immune cells is required for eradicating large intracranial tumors.


Assuntos
Neoplasias Encefálicas/imunologia , Glioma/imunologia , Microglia/imunologia , Neoplasias Experimentais/imunologia , Linfócitos T/imunologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Linhagem Celular Tumoral , Glioma/patologia , Rejeição de Enxerto/imunologia , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Ratos , Ratos Endogâmicos F344 , Ratos Wistar , Transplante Homólogo , Transplante Isogênico
15.
Glia ; 53(6): 669-74, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16419088

RESUMO

Microglial cells undergo cell division in vitro, as well as in vivo after brain injury. Mitotic activity of microglia suggests that they have limited life spans and rely on self-renewal to replace senescent cells. In the current study we examined long-term effects of antioxidants vitamin E and alpha-lipoic acid on cultured rat microglia with respect to proliferative ability, telomere length, telomerase activity, and interleukin-1beta (IL-1beta) production. We report that vitamin E induces dramatic microglial proliferation, as measured by MTT assay and BrdU incorporation, surpassing that of the well-known microglial mitogen granulocyte macrophage-colony stimulating factor, and therefore establishing vitamin E as the most potent, known mitogen for microglia in vitro. The high rate of microglial proliferation resulted in a concomitant decrease in telomere length and telomerase activity. Production of IL-1beta was significantly decreased in vitamin E-treated microglia in vitro. Our findings provide an impetus to investigate potential benefits of vitamin E supplementation on microglial renewal capacity in vivo during aging or after brain injury.


Assuntos
Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Proliferação de Células/efeitos dos fármacos , Gliose/metabolismo , Microglia/metabolismo , alfa-Tocoferol/farmacologia , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Encéfalo/citologia , Encéfalo/fisiopatologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/fisiopatologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Gliose/induzido quimicamente , Gliose/fisiopatologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-1/metabolismo , Microglia/efeitos dos fármacos , Mitógenos/metabolismo , Mitógenos/farmacologia , Mitógenos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Telomerase/antagonistas & inibidores , Telomerase/metabolismo , Telômero/efeitos dos fármacos , Telômero/fisiologia , Ácido Tióctico/metabolismo , Ácido Tióctico/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , alfa-Tocoferol/metabolismo , alfa-Tocoferol/uso terapêutico
16.
Neurol Res ; 27(7): 685-91, 2005 10.
Artigo em Inglês | MEDLINE | ID: mdl-16197805

RESUMO

Microglial cells comprise a network of endogenous immunocompetent cells that pervade the brain and spinal cord. The primary function of this system is to provide continuous surveillance of the parenchyma and protect the central nervous system (CNS) during injury and disease. Here we discuss the involvement of microglia during brain aging and aging-related neurodegenerative disease, i.e. Alzheimer's disease, and briefly summarize their possible roles in amyotrophic lateral sclerosis (ALS). In addition, we provide an overview of the neuroinflammation associated with primary brain tumors and how microglial tumor cytotoxicity could be targeted for immunotherapeutic approaches designed to treat these lesions.


Assuntos
Encéfalo/imunologia , Microglia/imunologia , Medula Espinal/imunologia , Doença de Alzheimer/imunologia , Animais , Neoplasias Encefálicas/imunologia , Humanos , Inflamação , Doença dos Neurônios Motores/imunologia
17.
Curr Alzheimer Res ; 2(2): 187-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15974917

RESUMO

Blocking the effects of fractalkine therapeutically may regulate microglia cell activation and provide neuroprotection in the AD brain. A human herpesvirus 8-encoded chemokine, termed vMIP-II is a non-selective chemokine receptor antagonist (binding multiple chemokine receptors, including CX3CR1). By comparing vMIP-II and FKN, we have generated molecules that selectively antagonize CX3CR1 activation. The results from these studies will guide future development of therapeutic agents designed to modulate microglial activation with the goal of preventing or slowing the progression of AD.


Assuntos
Comunicação Celular/fisiologia , Quimiocinas CX3C/antagonistas & inibidores , Herpesvirus Humano 8 , Proteínas de Membrana/antagonistas & inibidores , Microglia/fisiologia , Neurônios/fisiologia , Sequência de Aminoácidos/genética , Animais , Quimiocina CX3CL1 , Quimiocinas CX3C/fisiologia , Herpesvirus Humano 8/genética , Humanos , Proteínas de Membrana/fisiologia , Dados de Sequência Molecular
18.
Immunology ; 114(4): 565-74, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15804293

RESUMO

Stromal-derived factor-1 (SDF-1/CXCL12) and its receptor CXCR4 play crucial roles in leukocyte migration and activation, as well as embryogenesis, angiogenesis, cancer and viral pathogenesis. CXCR4 is one of the major human immunodeficiency virus-1 (HIV-1) coreceptors on macrophages. In many tissues macrophages are one of the predominant cell types infected by HIV-1 and act as a reservoir for persistent infection and viral dissemination. In patients infected by HIV-1, blood and tissue levels of transforming growth factor-beta1 (TGF-beta1) are increased. The purpose of this study was to evaluate the effects of TGF-beta1 on CXCR4 expression and function in primary human monocyte-derived macrophages (MDMs) and rat microglia. TGF-beta1 up-regulated CXCR4 and enhanced SDF-1alpha-stimulated ERK1,2 phosphorylation in these cells. The increased CXCR4 expression in human MDMs resulted in increased susceptibility of the cells to entry by dual-tropic CXCR4-using HIV-1 (D-X4). In contrast, TGF-beta1 failed to increase CCR5 expression or infection by a CCR5-using virus in MDMs. Our data demonstrate that TGF-beta1 enhances macrophage responsiveness to SDF-1alpha stimulation and susceptibility to HIV-1 by selectively increasing expression of CXCR4. The results suggest that increased expression of CXCR4 on macrophages may contribute to the emergence of dual-tropic X4 viral variants at later stages of HIV-1 infection.


Assuntos
Quimiocinas CXC/metabolismo , Infecções por HIV/imunologia , HIV-1 , Macrófagos/imunologia , Receptores de Quimiocinas/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Células Cultivadas , Quimiocina CXCL12 , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Infecções por HIV/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/virologia , Microglia , Microscopia de Fluorescência , Fosforilação , Ratos , Receptores CXCR4
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA