Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cancer Immunol Immunother ; 63(9): 911-24, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24893855

RESUMO

BACKGROUND: Cancer vaccines reproducibly cure laboratory animals and reveal encouraging trends in brain tumor (glioma) patients. Identifying parameters governing beneficial vaccine-induced responses may lead to the improvement of glioma immunotherapies. CD103(+) CD8 T cells dominate post-vaccine responses in human glioma patients for unknown reasons, but may be related to recent thymic emigrant (RTE) status. Importantly, CD8 RTE metrics correlated with beneficial immune responses in vaccinated glioma patients. METHODS: We show by flow cytometry that murine and human CD103(+) CD8 T cells respond better than their CD103(-) counterparts to tumor peptide-MHC I (pMHC I) stimulation in vitro and to tumor antigens on gliomas in vivo. RESULTS: Glioma responsive T cells from mice and humans both exhibited intrinsic de-sialylation-affecting CD8 beta. Modulation of CD8 T cell sialic acid with neuraminidase and ST3Gal-II revealed de-sialylation was necessary and sufficient for promiscuous binding to and stimulation by tumor pMHC I. Moreover, de-sialylated status was required for adoptive CD8 T cells and lymphocytes to decrease GL26 glioma invasiveness and increase host survival in vivo. Finally, increased tumor ST3Gal-II expression correlated with clinical vaccine failure in a meta-analysis of high-grade glioma patients. CONCLUSIONS: Taken together, these findings suggest that de-sialylation of CD8 is required for hyper-responsiveness and beneficial anti-glioma activity by CD8 T cells. Because CD8 de-sialylation can be induced with exogenous enzymes (and appears particularly scarce on human T cells), it represents a promising target for clinical glioma vaccine improvement.


Assuntos
Antígenos CD/imunologia , Neoplasias Encefálicas/terapia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/farmacologia , Células Dendríticas/imunologia , Glioma/terapia , Cadeias alfa de Integrinas/imunologia , Animais , Antígenos CD/metabolismo , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/imunologia , Feminino , Glioblastoma/imunologia , Glioblastoma/metabolismo , Glioblastoma/terapia , Glioma/imunologia , Glioma/metabolismo , Humanos , Imunoterapia Adotiva/métodos , Cadeias alfa de Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuraminidase/metabolismo , Neuraminidase/farmacologia , Sialiltransferases/metabolismo , Sialiltransferases/farmacologia , beta-Galactosídeo alfa-2,3-Sialiltransferase
2.
bioRxiv ; 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38328072

RESUMO

Cerebral (Aß) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aß/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aß/fibrillar pTau, however, appears to vary depending on the animal model used. Our prior work suggested that antigen-specific memory CD8 T (" hi T") cells act upstream of Aß/pTau after brain injury. Here we examine whether hi T cells influence sporadic AD-like pathophysiology upstream of Aß/pTau. Examining neuropathology, gene expression, and behavior in our hi T mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. Our work is the first to identify an age-related factor acting upstream of Aß/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD. Significance Statement: This study changes our view of Alzheimer's Disease (AD) initiation and progression. Mutations promoting cerebral beta-amyloid (Aß) deposition guarantee rare genetic forms of AD. Thus, the prevailing hypothesis has been that Aß is central to initiation and progression of all AD, despite contrary animal and patient evidence. We show that age-related T cells generate neurodegeneration with compelling features of AD in mice, with distinct T cell functions required for pathological initiation and neurodegenerative progression. Knowledge from these mice was applied to successfully predict previously unknown features of human AD and generate novel tools for its clinical management.

3.
Front Neurol ; 11: 557269, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424735

RESUMO

The incidence of autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD), which frequently co-occur, are both rising. The causes of ASD and ADHD remain elusive, even as both appear to involve perturbation of the gut-brain-immune axis. CD103 is an integrin and E-cadherin receptor most prominently expressed on CD8 T cells that reside in gut, brain, and other tissues. CD103 deficiency is well-known to impair gut immunity and resident T cell function, but it's impact on neurodevelopmental disorders has not been examined. We show here that CD8 T cells influence neural progenitor cell function, and that CD103 modulates this impact both directly and potentially by controlling CD8 levels in brain. CD103 knockout (CD103KO) mice exhibited a variety of behavioral abnormalities, including superior cognitive performance coupled with repetitive behavior, aversion to novelty and social impairment in females, with hyperactivity with delayed learning in males. Brain protein markers in female and male CD103KOs coincided with known aspects of ASD and ADHD in humans, respectively. Surprisingly, CD103 deficiency also decreased age-related cognitive decline in both sexes, albeit by distinct means. Together, our findings reveal a novel role for CD103 in brain developmental function, and identify it as a unique factor linking ASD and ADHD etiology. Our data also introduce a new animal model of combined ASD and ADHD with associated cognitive benefits, and reveal potential therapeutic targets for these disorders and age-related cognitive decline.

4.
Clin Cancer Res ; 14(12): 4002-9, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559623

RESUMO

PURPOSE: The blood-brain tumor barrier (BTB) significantly limits the delivery of chemotherapeutics to brain tumors. Nitric oxide (NO) is involved in the regulation of cerebral vascular permeability. We investigated the effects of NO donors, L-arginine and hydroxyurea, on BTB permeability in 9L gliosarcoma-bearing Fischer rats. EXPERIMENTAL DESIGN: The rats implanted with 9L gliosarcoma were dosed orally with hydroxyurea and L-arginine. BTB permeability, defined by the unidirectional transport constant, Ki, for [14C]sucrose was measured. The expression of neural and endothelial NO synthase (NOS) in tumors and normal brain tissue was examined. Further, the levels of NO, L-citrulline, and cGMP in the tumor and normal brain tissue were measured. RESULTS: Oral administration of l-arginine or hydroxyurea significantly increased BTB permeability when compared with the nontreated control. The selective effects were abolished by iberiotoxin, an antagonist of calcium-dependent potassium (KCa) channel that is a cGMP pathway effector. The expression of endothelial NOS, but not neural NOS, was higher in tumor vessels than in those of normal brain. Moreover, the levels of NO, L-citrulline, a byproduct of NO formation from L-arginine, and cGMP were enhanced in the tumor tissue by oral administration of L-arginine and/or hydroxyurea. CONCLUSIONS: Oral administration of L-arginine or hydroxyurea selectively increased tumor permeability, which is likely mediated by alteration in cGMP levels. The findings suggest that use of oral NO donors may be a strategy to enhance the delivery of chemotherapeutics to malignant brain tumors.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Permeabilidade Capilar/efeitos dos fármacos , Glioma/patologia , Doadores de Óxido Nítrico/farmacologia , Administração Oral , Animais , Arginina/administração & dosagem , Arginina/farmacologia , Barreira Hematoencefálica/fisiologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatologia , Permeabilidade Capilar/fisiologia , Citrulina/metabolismo , GMP Cíclico/metabolismo , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Glioma/metabolismo , Glioma/fisiopatologia , Hidroxiureia/administração & dosagem , Hidroxiureia/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Ratos , Ratos Endogâmicos F344 , Células Tumorais Cultivadas
5.
Neurobiol Dis ; 30(3): 343-352, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18417351

RESUMO

The degeneration of neurons in the mammalian brain is commonly associated with the division of cells located in the damaged area. The aim of the present study has been to characterise the phenotype of newly born cells in the striatum of adult rats following 6-hydroxydopamine lesion of the nigro-striatal pathway. Newborn cells were identified through labelling with either bromodeoxyuridine or retrovirus encoding green fluorescence protein. We report here that the overwhelming majority of these cells have glial characteristics. In order to promote the generation of new neurons we retrovirally introduced either the noggin or neurogenin2 genes into newborn cells following the 6-hydroxydopamine lesion. Transduction with neurogenin2 resulted in the production of cells resembling neuroblasts, however these cells did not appear to survive. Noggin transduction did not result in the generation of new neurons, but interestingly, greatly increased the number of oligodendrocytes generated from newborn cells.


Assuntos
Proliferação de Células , Corpo Estriado/fisiologia , Técnicas de Transferência de Genes , Oxidopamina/toxicidade , Substância Negra/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Feminino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Substância Negra/citologia , Substância Negra/efeitos dos fármacos
6.
Brain Res ; 1227: 198-206, 2008 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-18602898

RESUMO

The blood-brain tumor barrier (BTB) significantly limits delivery of effective concentrations of chemotherapeutic drugs to brain tumors. Previous studies suggest that BTB permeability may be modulated via alteration in the activity of potassium channels. In this study, we studied the relationship of BTB permeability increase mediated by potassium channel agonists to channel expression in two rat brain tumor models. Intravenous infusion of KCO912 (K(ATP) agonist), minoxidil sulfate (K(ATP) agonist) or NS1619 (K(Ca) agonist) increased tumor permeability more in the 9L allogeneic brain tumor model than in the syngeneic brain tumor model. Consistently, expression of both K(ATP) and K(Ca) channels in 9L tumors was increased to a significantly greater extent in Wistar rats (allogeneic) as compared to Fischer rats (syngeneic). Furthermore, as a preliminary effort to understand clinical implication of potassium channels in brain tumor treatment, we determined the expression of K(ATP) in surgical specimens. K(ATP) mRNA was detected in glioblastoma multiforme (GBM) from nineteen patients examined, with a wide range of expression levels. Interestingly, in paired GBM tissues from seven patients before and after vaccination therapy, increased levels of K(ATP) were detected in five patients after vaccination that had positive response to chemotherapy after vaccination. The present study indicates that the effects of potassium channel agonists on BTB permeability are different between syngeneic and allogeneic models which have different expression levels of potassium channels. The expression of potassium channels in brain tumors is variable, which may be associated with different tumor permeability to therapeutic agents among patients.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Permeabilidade Capilar/efeitos dos fármacos , Canais KATP/agonistas , Canais de Potássio Cálcio-Ativados/agonistas , Animais , Benzimidazóis/administração & dosagem , Benzimidazóis/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Western Blotting , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Feminino , Glioblastoma/patologia , Glioblastoma/fisiopatologia , Glioblastoma/terapia , Humanos , Imuno-Histoquímica , Injeções Intravenosas , Canais KATP/genética , Canais KATP/fisiologia , Microscopia Confocal , Minoxidil/administração & dosagem , Minoxidil/análogos & derivados , Minoxidil/uso terapêutico , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Neoplasias Experimentais/fisiopatologia , Neoplasias Experimentais/terapia , Canais de Potássio Cálcio-Ativados/genética , Canais de Potássio Cálcio-Ativados/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Vacinação/métodos
7.
Brain Res ; 1230: 290-302, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18674521

RESUMO

The blood-brain tumor barrier (BTB) significantly limits delivery of therapeutic concentrations of chemotherapy to brain tumors. A novel approach to selectively increase drug delivery is pharmacologic modulation of signaling molecules that regulate BTB permeability, such as those in cGMP signaling. Here we show that oral administration of sildenafil (Viagra) and vardenafil (Levitra), inhibitors of cGMP-specific PDE5, selectively increased tumor capillary permeability in 9L gliosarcoma-bearing rats with no significant increase in normal brain capillaries. Tumor-bearing rats treated with the chemotherapy agent, adriamycin, in combination with vardenafil survived significantly longer than rats treated with adriamycin alone. The selective increase in tumor capillary permeability appears to be mediated by a selective increase in tumor cGMP levels and increased vesicular transport through tumor capillaries, and could be attenuated by iberiotoxin, a selective inhibitor for calcium-dependent potassium (K(Ca)) channels, that are effectors in cGMP signaling. The effect by sildenafil could be further increased by simultaneously using another BTB "opener", bradykinin. Collectively, this data demonstrates that oral administration of PDE5 inhibitors selectively increases BTB permeability and enhances anti-tumor efficacy for a chemotherapeutic agent. These findings have significant implications for improving delivery of anti-tumor agents to brain tumors.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Glioma/tratamento farmacológico , Glioma/metabolismo , Inibidores da Fosfodiesterase 5 , Inibidores de Fosfodiesterase/farmacologia , Animais , Autorradiografia , Pressão Sanguínea/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Capilares/patologia , Permeabilidade Capilar/efeitos dos fármacos , GMP Cíclico/sangue , GMP Cíclico/metabolismo , Feminino , Glioma/patologia , Imidazóis/farmacologia , Microscopia Eletrônica de Transmissão , Neovascularização Patológica/patologia , Piperazinas/farmacologia , Purinas/farmacologia , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Citrato de Sildenafila , Sacarose/metabolismo , Sulfonas/farmacologia , Análise de Sobrevida , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/ultraestrutura , Triazinas/farmacologia , Dicloridrato de Vardenafila
8.
Mol Cancer ; 6: 22, 2007 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-17359538

RESUMO

BACKGROUND: The blood-brain tumor barrier (BTB) impedes the delivery of therapeutic agents to brain tumors. While adequate delivery of drugs occurs in systemic tumors, the BTB limits delivery of anti-tumor agents into brain metastases. RESULTS: In this study, we examined the function and regulation of calcium-activated potassium (KCa) channels in a rat metastatic brain tumor model. We showed that intravenous infusion of NS1619, a KCa channel agonist, and bradykinin selectively enhanced BTB permeability in brain tumors, but not in normal brain. Iberiotoxin, a KCa channel antagonist, significantly attenuated NS1619-induced BTB permeability increase. We found KCa channels and bradykinin type 2 receptors (B2R) expressed in cultured human metastatic brain tumor cells (CRL-5904, non-small cell lung cancer, metastasized to brain), human brain microvessel endothelial cells (HBMEC) and human lung cancer brain metastasis tissues. Potentiometric assays demonstrated the activity of KCa channels in metastatic brain tumor cells and HBMEC. Furthermore, we detected higher expression of KCa channels in the metastatic brain tumor tissue and tumor capillary endothelia as compared to normal brain tissue. Co-culture of metastatic brain tumor cells and brain microvessel endothelial cells showed an upregulation of KCa channels, which may contribute to the overexpression of KCa channels in tumor microvessels and selectivity of BTB opening. CONCLUSION: These findings suggest that KCa channels in metastatic brain tumors may serve as an effective target for biochemical modulation of BTB permeability to enhance selective delivery of chemotherapeutic drugs to metastatic brain tumors.


Assuntos
Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Canais de Potássio Cálcio-Ativados/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Técnicas de Cocultura , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Permeabilidade/efeitos dos fármacos , Canais de Potássio Cálcio-Ativados/agonistas , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Canais de Potássio Cálcio-Ativados/genética , Potenciometria , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptor B2 da Bradicinina/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Mol Cancer ; 5: 67, 2006 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-17140455

RESUMO

BACKGROUND: Recently, a small population of cancer stem cells in adult and pediatric brain tumors has been identified. Some evidence has suggested that CD133 is a marker for a subset of leukemia and glioblastoma cancer stem cells. Especially, CD133 positive cells isolated from human glioblastoma may initiate tumors and represent novel targets for therapeutics. The gene expression and the drug resistance property of CD133 positive cancer stem cells, however, are still unknown. RESULTS: In this study, by FACS analysis we determined the percentage of CD133 positive cells in three primary cultured cell lines established from glioblastoma patients 10.2%, 69.7% and 27.5%, respectively. We also determined the average mRNA levels of markers associated with neural precursors. For example, CD90, CD44, CXCR4, Nestin, Msi1 and MELK mRNA on CD133 positive cells increased to 15.6, 5.7, 337.8, 21.4, 84 and 1351 times, respectively, compared to autologous CD133 negative cells derived from cell line No. 66. Additionally, CD133 positive cells express higher levels of BCRP1 and MGMT mRNA, as well as higher mRNA levels of genes that inhibit apoptosis. Furthermore, CD133 positive cells were significantly resistant to chemotherapeutic agents including temozolomide, carboplatin, paclitaxel (Taxol) and etoposide (VP16) compared to autologous CD133 negative cells. Finally, CD133 expression was significantly higher in recurrent GBM tissue obtained from five patients as compared to their respective newly diagnosed tumors. CONCLUSION: Our study for the first time provided evidence that CD133 positive cancer stem cells display strong capability on tumor's resistance to chemotherapy. This resistance is probably contributed by the CD133 positive cell with higher expression of on BCRP1 and MGMT, as well as the anti-apoptosis protein and inhibitors of apoptosis protein families. Future treatment should target this small population of CD133 positive cancer stem cells in tumors to improve the survival of brain tumor patients.


Assuntos
Antígenos CD/biossíntese , Antineoplásicos/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glicoproteínas/biossíntese , Células-Tronco Neoplásicas/fisiologia , Antígeno AC133 , Antígenos CD/genética , Apoptose/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Expressão Gênica , Glioblastoma/imunologia , Glioblastoma/patologia , Glicoproteínas/genética , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/imunologia , Peptídeos/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
10.
Neurosurg Focus ; 20(4): E27, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16709033

RESUMO

The observation of similarities between the self-renewal mechanisms of stem cells and cancer cells has led to the new concept of the cancer stem cell. In cases of leukemia, multiple myeloma, and breast cancer, cells with a high selfrenewal potential have been identified. Furthermore, investigators have shown these cells' ability to drive the formation and growth of the tumor. Brain tumors have also been reported to possess a subpopulation of cancer stemlike cells that have the ability to proliferate, self-renew, and be multipotent. When grafted into mice, these cells are also able to generate a tumor that recapitulates that of the patient from whom the cells were derived. The identification and characterization of this new category of cells call for new therapies capable of selectively targeting and killing these multifaceted cells.


Assuntos
Neoplasias Encefálicas/imunologia , Transformação Celular Neoplásica/imunologia , Imunoterapia/métodos , Células-Tronco Multipotentes/imunologia , Células-Tronco Neoplásicas/imunologia , Antígeno AC133 , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/imunologia , Animais , Antígenos CD/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/síntese química , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Divisão Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Glicoproteínas/imunologia , Humanos , Imunoterapia/tendências , Camundongos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/transplante , Proteínas de Neoplasias/imunologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/transplante , Peptídeos/imunologia
11.
J Neurosci ; 23(3): 748-57, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12574403

RESUMO

RAS proteins are critical regulators of mitosis and are mutationally activated in many human tumors. RAS signaling is also known to mediate long-term potentiation (LTP) and long-term memory formation in postmitotic neurons, in part through activation of the RAF-MEK-ERK pathway. The RAS effector RIN1 appears to function through competitive inhibition of RAS-RAF binding and also through diversion of RAS signaling to alternate pathways. We show that RIN1 is preferentially expressed in postnatal forebrain neurons in which it is localized in dendrites and physically associated with RAS, suggesting a role in RAS-mediated postsynaptic neuronal plasticity. Mice with an Rin1 gene disruption showed a striking enhancement in amygdala LTP. In addition, two independent behavioral tests demonstrated elevated amygdala-dependent aversive memory in Rin1(-/-) mice. These results indicate that RIN1 serves as an inhibitory modulator of neuronal plasticity in aversive memory formation.


Assuntos
Proteínas de Transporte/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Proteínas rab de Ligação ao GTP , Proteínas ras/metabolismo , Tonsila do Cerebelo/fisiologia , Animais , Proteínas de Transporte/genética , Contagem de Células , Condicionamento Clássico , Dendritos/metabolismo , Medo/fisiologia , Viabilidade Fetal/genética , Hipocampo/fisiologia , Humanos , Aprendizagem/fisiologia , Potenciação de Longa Duração/genética , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/ultraestrutura , Prosencéfalo/química , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Ligação Proteica/fisiologia
12.
J Neurol Neurophysiol ; 5(3)2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-25346865

RESUMO

T-lymphocytes have been previously implicated in protecting dopaminergic neurons in the substantianigra from induced cell death. However, the role of T-cells in neurodegenerative models such as Parkinson's disease (PD) has not been fully elucidated. To examine the role of T-lymphocytes on motor behavior in the 6-hydroxydopamine (6-OHDA) unilateral striatal partial lesion PD rat model, we assessed progression of hemi-parkinsonian lesions in the substantia nigra, induced by 6-OHDA striatal injections, in athymic rats (RNU-/-, T-lymphocyte-deficient) as compared to RNU-/+ rats (phenotypically normal). Motor skills were determined by the cylinder and D-amphetamine sulfate-induced rotational behavioral tests. Cylinder behavioral test showed no significant difference between unilaterally lesioned RNU-/- and RNU-/+ rats. However both unilaterally lesioned RNU-/- and RNU-/+ rats favored the use of the limb ipsilateral to lesion. Additionally, amphetamine-induced rotational test revealed greater rotational asymmetry in RNU-/- rats compared to RNU-/+ rats at two- and six-week post-lesion. Quantitative immunohistochemistry confirmed loss of striatal TH-immunopositive fibers in RNU-/- and RNU-/+ rat, as well as blood-brain-barrier changes associated with PD that may influence passage of immune cells into the central nervous system in RNU-/- brains. Specifically, GFAP immunopositive cells were decreased, as were astrocytic end-feet (AQP4) contacting blood vessels (laminin) in the lesioned relative to contralateral striatum. Flow cytometric analysis in 6-OHDA lesioned RNU-/+rats revealed increased CD4+ and decreased CD8+ T cells specifically within lesioned brain. These results suggest that both major T cell subpopulations are significantly and reciprocally altered following 6-OHDA-lesioning, and that global T cell deficiency exacerbates motor behavioral defects in this rat model of PD.

13.
J Neurosurg ; 117(4): 705-11, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22920963

RESUMO

OBJECT: The prognosis of patients with glioblastoma who present with multifocal disease is not well documented. The objective of this study was to determine whether multifocal disease on initial presentation is associated with worse survival. METHODS: The authors retrospectively reviewed records of 368 patients with newly diagnosed glioblastoma and identified 47 patients with multifocal tumors. Each patient with a multifocal tumor was then matched with a patient with a solitary glioblastoma on the basis of age, Karnofsky Performance Scale (KPS) score, and extent of resection, using a propensity score matching methodology. Radiation and temozolomide treatments were also well matched between the 2 cohorts. Kaplan-Meier estimates and log-rank tests were used to compare patient survival. RESULTS: The incidence of multifocal tumors was 12.8% (47/368). The median age of patients with multifocal tumors was 61 years, 76.6% had KPS scores ≥ 70, and 87.2% underwent either a biopsy or partial resection of their tumors. The 47 patients with multifocal tumors were almost perfectly matched on the basis of age (p = 0.97), extent of resection (p = 1.0), and KPS score (p = 0.80) compared with 47 patients with a solitary glioblastoma. Age (>65 years), partial resection or biopsy, and low KPS score (<70) were associated with worse median survival within the multifocal group. In the multifocal group, 19 patients experienced tumor progression on postradiation therapy MRI, compared with 11 patients (26.8%) with tumor progression in the unifocal group (p = 0.08). Patients with multifocal tumors experienced a significantly shorter median overall survival of 6 months (95% CI 4-10 months), compared with the 11-month median survival (95% CI 10-19 months) of the matched solitary glioblastoma group (p = 0.02, log-rank test). Two-year survival rates were 4.3% for patients with multifocal tumors and 29.0% for the unifocal cohort. Patients with newly diagnosed multifocal tumors were found to have an almost 2-fold increase in the hazard of death compared with patients with solitary glioblastoma (hazard ratio 1.8, 95% CI 1.1-3.1; p = 0.02). Tumor samples were analyzed for expression of phosphorylated mitogen-activated protein kinase, phosphatase and tensin homolog, O(6)-methylguanine-DNA methyltransferase, laminin ß1 and ß2, as well as epidermal growth factor receptor amplification, and no significant differences in expression profile between the multifocal and solitary glioblastoma groups was found. CONCLUSIONS: Patients with newly diagnosed multifocal glioblastoma on presentation experience significantly worse survival than patients with solitary glioblastoma. Patients with multifocal tumors continue to pose a therapeutic challenge in the temozolomide era and magnify the challenges faced while treating patients with malignant gliomas.


Assuntos
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Glioblastoma/diagnóstico , Glioblastoma/terapia , Neoplasias Primárias Múltiplas/diagnóstico , Neoplasias Primárias Múltiplas/terapia , Idoso , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/mortalidade , Estudos de Casos e Controles , Estudos de Coortes , Terapia Combinada , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Glioblastoma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Neoplasias Primárias Múltiplas/mortalidade , Procedimentos Neurocirúrgicos , Prognóstico , Radioterapia , Estudos Retrospectivos , Taxa de Sobrevida , Temozolomida , Resultado do Tratamento
14.
PLoS One ; 5(6): e10974, 2010 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-20539758

RESUMO

BACKGROUND: Small populations of highly tumorigenic stem-like cells (cancer stem cells; CSCs) can exist within, and uniquely regenerate cancers including malignant brain tumors (gliomas). Many aspects of glioma CSCs (GSCs), however, have been characterized in non-physiological settings. METHODS: We found gene expression similarity superiorly defined glioma "stemness", and revealed that GSC similarity increased with lower tumor grade. Using this method, we examined stemness in human grade IV gliomas (GBM) before and after dendritic cell (DC) vaccine therapy. This was followed by gene expression, phenotypic and functional analysis of murine GL26 tumors recovered from nude, wild-type, or DC-vaccinated host brains. RESULTS: GSC similarity was specifically increased in post-vaccine GBMs, and correlated best to vaccine-altered gene expression and endogenous anti-tumor T cell activity. GL26 analysis confirmed immune alterations, specific acquisition of stem cell markers, specifically enhanced sensitivity to anti-stem drug (cyclopamine), and enhanced tumorigenicity in wild-type hosts, in tumors in proportion to anti-tumor T cell activity. Nevertheless, vaccine-exposed GL26 cells were no more tumorigenic than parental GL26 in T cell-deficient hosts, though they otherwise appeared similar to GSCs enriched by chemotherapy. Finally, vaccine-exposed GBM and GL26 exhibited relatively homogeneous expression of genes expressed in progenitor cells and/or differentiation. CONCLUSIONS: T cell activity represents an inducible physiological process capable of proportionally enriching GSCs in human and mouse gliomas. Stem-like gliomas enriched by strong T cell activity, however, may differ from other GSCs in that their stem-like properties may be disassociated from increased tumor malignancy and heterogeneity under specific host immune conditions.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Células-Tronco Neoplásicas/citologia , Linfócitos T Citotóxicos/citologia , Animais , Sequência de Bases , Neoplasias Encefálicas/genética , Primers do DNA , Citometria de Fluxo , Imunofluorescência , Glioma/genética , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase
15.
Cancer Res ; 68(14): 5955-64, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18632651

RESUMO

Cancer vaccine trials have failed to yield robust immune-correlated clinical improvements as observed in animal models, fueling controversy over the utility of human cancer vaccines. Therapeutic vaccination represents an intriguing additional therapy for glioblastoma multiforme (GBM; grade 4 glioma), which has a dismal prognosis and treatment response, but only early phase I vaccine trial results have been reported. Immune and clinical responses from a phase II GBM vaccine trial are reported here. IFN-gamma responsiveness was quantified in peripheral blood of 32 GBM patients given therapeutic dendritic cell vaccines. Posttreatment times to tumor progression (TTP) and survival (TTS) were compared in vaccine responders and nonresponders and were correlated with immune response magnitudes. GBM patients (53%) exhibited >or=1.5-fold vaccine-enhanced cytokine responses. Endogenous antitumor responses of similar magnitude occurred in 22% of GBM patients before vaccination. Vaccine responders exhibited significantly longer TTS and TTP relative to nonresponders. Immune enhancement in vaccine responders correlated logarithmically with TTS and TTP spanning postvaccine chemotherapy, but not with initial TTP spanning vaccination alone. This is the first report of a progressive correlation between cancer clinical outcome and T-cell responsiveness after therapeutic vaccination in humans and the first tracing of such correlation to therapeutically exploitable tumor alteration. As such, our findings offer unique opportunities to identify cellular and molecular components of clinically meaningful antitumor immunity in humans.


Assuntos
Neoplasias Encefálicas/microbiologia , Neoplasias Encefálicas/terapia , Glioblastoma/imunologia , Glioblastoma/terapia , Adulto , Idoso , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer , Células Dendríticas/imunologia , Feminino , Humanos , Sistema Imunitário , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Resultado do Tratamento
16.
Exp Cell Res ; 313(6): 1172-80, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17291494

RESUMO

Parkinson's Disease (PD) is a debilitating motor function disorder due primarily to a loss of midbrain dopaminergic neurons and a subsequent reduction in dopaminergic innervation of the striatum. Several attempts have been made to generate dopaminergic neurons from progenitor cell populations in vitro for potential use in cell replacement therapy for PD. However, expanding cells from fetal brain with retained potential for dopaminergic differentiation has proven to be difficult. In this study, we sought to generate mesencephalic dopaminergic (mesDA) neurons from an expanded population of fetal mouse ventral midbrain (VM) progenitors through the use of retroviral gene delivery. We over-expressed Ngn2 and Nurr1, two genes present in the ventral midbrain and important for normal development of mesDA neurons, in multi-passaged neurosphere-expanded midbrain progenitors. We show that over-expression of Ngn2 in these progenitors results in increased neuronal differentiation but does not promote mesDA formation. We also show that over-expression of Nurr1 alone is sufficient to generate tyrosine hydroxylase (TH) expressing cells with an immature morphology, however the cells do not express any additional markers of mesDA neurons. Over-expression of Nurr1 and Ngn2 in combination generates morphologically mature TH-expressing neurons that also express additional mesencephalic markers.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Ligação a DNA/fisiologia , Dopamina/metabolismo , Mesencéfalo/citologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular , Mesencéfalo/metabolismo , Camundongos , Neurônios/enzimologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , Células-Tronco/enzimologia , Tirosina 3-Mono-Oxigenase/metabolismo
17.
Stem Cells ; 25(7): 1645-53, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17412894

RESUMO

The rat 9L gliosarcoma is a widely used syngeneic rat brain tumor model that closely simulates glioblastoma multiforme when implanted in vivo. In this study, we sought to isolate and characterize a subgroup of cancer stem-like cells (CSLCs) from the 9L gliosarcoma cell line, which may represent the tumor-initiating subpopulation of cells. We demonstrate that these CSLCs form clonal-derived spheres in media devoid of serum supplemented with the mitogens epidermal growth factor and basic fibroblast growth factor, express the NSC markers Nestin and Sox2, self-renew, and differentiate into neuron-like and glial cells in vitro. More importantly, these cells can propagate and recapitulate tumors when implanted into the brain of syngeneic Fisher rats, and they display a more aggressive course compared with 9L gliosarcoma cells grown in monolayer cultures devoid of mitogens. Furthermore, we compare the chemosensitivity and proliferation rate of 9L gliosarcoma cells grown as a monolayer to those of cells grown as floating spheres and show that the sphere-generated cells have a lower proliferation rate, are more chemoresistant, and express several antiapoptosis and drug-related genes, which may prove to have important clinical implications. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Gliossarcoma/patologia , Células-Tronco Neoplásicas/patologia , Esferoides Celulares/patologia , Animais , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Separação Celular , Medições Luminescentes , Células-Tronco Neoplásicas/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/metabolismo , Ratos , Esferoides Celulares/efeitos dos fármacos , Análise de Sobrevida , Células Tumorais Cultivadas
18.
J Neurosci Res ; 75(3): 330-43, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14743446

RESUMO

The Notch-DSL signaling system, consisting of multiple receptors and ligands, inhibits neurogenesis and promotes gliogenesis during embryonic development, but the specific function of the various ligands and receptors at later developmental stages are unknown. Here, we examined the expression pattern of four Delta, Serrate and Lag-2 (DSL) ligands, Jagged1, Jagged2, Delta-like1 (Dl1) and Delta-like 3 (Dl3), in late embryonic and postnatal rat brain by in situ hybridization. In late embryos, Jagged1, Dl1 and Dl3 mRNAs were present in the periventricular germinal epithelia, but this expression diminished during postnatal ages. Jagged1 mRNA was also expressed in the inner aspect of the dentate gyrus at early postnatal times. Dl3 was detectable in the external granule cell layer (EGL) of the cerebellum, another site of postnatal neurogenesis. Jagged2 mRNA was expressed in virtually all postnatal neurons. Jagged1 mRNA was highly expressed in several brain nuclei during postnatal development, with lower levels of expression in other grey matter regions. In white matter, Dl1 and Dl3 mRNAs were expressed during the first week of postnatal development but only the expression of Dl1 mRNA persisted through the second week. Dl1 mRNA was present at lower levels throughout grey matter during the first few weeks of development. Jagged1 mRNA was expressed in blood vessels, choroid plexus, and menninges throughout development and in the adult. Jagged2 mRNA was transiently expressed in cerebral blood vessels and choroid plexus during the first postnatal week. Taken together, these results support multiple and differing roles for the various ligands during and after central nervous system (CNS) development.


Assuntos
Encéfalo/crescimento & desenvolvimento , Proteínas de Transporte/biossíntese , Proteínas de Membrana/biossíntese , Biossíntese de Proteínas , Proteínas , Animais , Animais Recém-Nascidos , Encéfalo/irrigação sanguínea , Encéfalo/embriologia , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Embrião de Mamíferos , Desenvolvimento Embrionário e Fetal , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Proteína Jagged-1 , Proteína Jagged-2 , Masculino , Neurônios/metabolismo , RNA Mensageiro , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Serrate-Jagged , Células-Tronco/metabolismo
19.
Dev Neurosci ; 25(2-4): 162-72, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12966214

RESUMO

Central nervous system germinal zones contain stem cells that generate both neurons and glia. In the recent past, these cells have been isolated, maintained in a variety of culture systems and used in vitro for subsequent characterization of molecular mechanisms underlying brain development. Factors that govern cell fate choices of these neural stem cells have not been fully elucidated, but recent studies suggest that age at the time of culture is an important intrinsic mechanism. Stem cell mitogens and Notch-DSL signaling are significant extrinsic factors. In the current study, we compare neurosphere cultures propagated from animals on embryonic day 12, embryonic day 18 and the day of birth and stimulated to divide by either basic fibroblast growth factor (bFGF) or transforming growth factor-alpha (TGF-alpha). As described for other systems, when bFGF was used, clonal neurospheres derived from the youngest age gave rise to a greater percentage of neurons. When TGF-alpha, acting via the epidermal growth factor receptor, was used, this effect was not observed, with neurospheres from younger animals giving rise to a similar percentage of neurons as those derived from older animals suggesting that this growth factor was either stimulating a different population of stem cells to proliferate, or that it was capable of overriding intrinsic mechanisms. Other differences were also observed when the two growth factors were compared, including age-dependent differences in the numbers of putative astrocytes and oligodendrocytes formed. We further assessed age-dependent influences on cell fate by assessing the effects of a lentivirally transduced constitutively activated Notch receptor on cell fate. At all ages studied, Notch activation resulted in a significantly greater number of GFAP-positive cells, seemingly overriding the greater neurogenic potential of younger stem cells. These data suggest that both extrinsic and intrinsic factors differentially regulate cell fate choices of progenitors during cortical development.


Assuntos
Envelhecimento/fisiologia , Diferenciação Celular/fisiologia , Córtex Cerebral/embriologia , Neurônios/citologia , Células-Tronco/citologia , Animais , Astrócitos/citologia , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Embrião de Mamíferos , Desenvolvimento Embrionário e Fetal/fisiologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Imunofluorescência , Vetores Genéticos , Humanos , Lentivirus/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Receptores Notch , Células-Tronco/efeitos dos fármacos , Transdução Genética , Fator de Crescimento Transformador alfa/farmacologia
20.
Dev Biol ; 264(2): 309-22, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14651920

RESUMO

The identification of the genes regulating neural progenitor cell (NPC) functions is of great importance to developmental neuroscience and neural repair. Previously, we combined genetic subtraction and microarray analysis to identify genes enriched in neural progenitor cultures. Here, we apply a strategy to further stratify the neural progenitor genes. In situ hybridization demonstrates expression in the central nervous system germinal zones of 54 clones so identified, making them highly relevant for study in brain and neural progenitor development. Using microarray analysis we find 73 genes enriched in three neural stem cell (NSC)-containing populations generated under different conditions. We use the custom microarray to identify 38 "stemness" genes, with enriched expression in the three NSC conditions and present in both embryonic stem cells and hematopoietic stem cells. However, comparison of expression profiles from these stem cell populations indicates that while there is shared gene expression, the amount of genetic overlap is no more than what would be expected by chance, indicating that different stem cells have largely different gene expression patterns. Taken together, these studies identify many genes not previously associated with neural progenitor cell biology and also provide a rational scheme for stratification of microarray data for functional analysis.


Assuntos
Encéfalo/metabolismo , Embrião de Mamíferos/embriologia , Perfilação da Expressão Gênica , Neurônios/citologia , Células-Tronco/metabolismo , Animais , Encéfalo/citologia , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Hibridização In Situ , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA