Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lab Anim (NY) ; 49(8): 227-232, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32690932

RESUMO

Despite several therapeutics showing promise in nonclinical studies, survival from ovarian cancer remains poor. New technologies are urgently needed to optimize the translation of nonclinical studies into clinical successes. While most nonclinical settings utilize subjective measures of physiological parameters, which can hamper the accuracy of the results, this study assessed the physical activity of mice in real time using an objective, non-invasive, cloud-based, digital vivarium monitoring platform. An initial range-finding study in which varying numbers of ovarian cancer cells were inoculated in mice was conducted to characterize disease progression using digital metrics such as motion and breathing rate. Data from the range-finding study were used to establish a motion threshold (MT) that might predict terminal endpoint. Using the MT, the efficacies of cisplatin and OS2966, an anti-CD29 antibody, were assessed. Results showed that MT predicted terminal endpoint significantly earlier than traditional parameters and correlated with therapeutic efficacy. Thus, continuous motion monitoring sensitively predicts terminal endpoint in nonclinical ovarian cancer models and could be applicable for drug efficacy testing.


Assuntos
Benchmarking , Neoplasias Ovarianas , Animais , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Camundongos , Neoplasias Ovarianas/tratamento farmacológico
2.
Target Oncol ; 14(4): 479-489, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31301014

RESUMO

BACKGROUND: High-grade meningiomas (HGMs; World Health Organization [WHO] classification grade II and III) have high relapse rates and poor clinical outcomes despite surgery and radiation treatments. No effective medical therapy currently exists for HGMs, and developing novel therapeutic strategies depends on the identification of molecular drivers. In cancer, ß1 integrin enhances malignant characteristics, including proliferation, invasion, and drug resistance. OBJECTIVE: We conducted this study to investigate whether ß1 integrin could be a therapeutic target in HGMs. PATIENTS AND METHODS: Expression of ß1 integrin was examined in gene array datasets, with proteomics of clinical meningioma specimens, and in patient-derived HGM xenografts. Anti-tumor activity of OS2966, a first-in-class humanized antagonizing monoclonal antibody against ß1 integrin, was tested in vitro and in vivo using an orthotopic mouse model of patient-derived malignant meningioma. RESULTS: ß1 integrin was expressed in meningiomas of all WHO grades and two xenografts tested. In vitro, OS2966 suppressed the viability of NF2-deficient MN3 sphere cells and NF2-wild-type IOMM-Lee malignant meningioma cells only when plated on laminin-coated plastic. While OS2966 decreased phosphorylation of ERK1/2 in both MN3 cells and laminin-grown IOMM-Lee cells, OS2966 only affected the phosphorylation of FAK (Tyr397) in MN3, and of Akt (Ser473) in IOMM-Lee cells, respectively, indicating differential pathway inhibition. Systemic administration of OS2966 in mice bearing orthotopic MN3 HGMs inhibited HGM cell proliferation and significantly extended overall survival of the treated mice. CONCLUSIONS: ß1 Integrin may be a therapeutic target in HGMs, and further preclinical and clinical development of OS2966 for HGM therapy is warranted.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Integrina beta1/genética , Meningioma/tratamento farmacológico , Meningioma/genética , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Proliferação de Células , Feminino , Humanos , Meningioma/patologia , Camundongos , Gradação de Tumores
3.
Mol Cancer Ther ; 18(6): 1127-1136, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30926634

RESUMO

Integrin ß1 receptor, expressed on the surface of tumor cells and macrophages in the tumor microenvironment (TME), has been implicated in both tumor progression and resistance to multiple modalities of therapy. OS2966 is the first clinical-ready humanized monoclonal antibody to block integrin ß1 and was recently orphan designated by the FDA Office of Orphan Products Development. Here, we tested therapeutic potential of OS2966-mediated integrin ß1 blockade to enhance the efficacy of oncolytic herpes simplex virus-1 (oHSV) through evaluation of virus replication, tumor cell killing efficiency, effect on the antiviral signaling pathway, co-culture assays of oHSV-infected cells with macrophages, and in vivo bioluminescence imaging on mammary fat pad triple-negative breast cancer xenograft and subcutaneous and intracranial glioma xenografts. OS2966 treatment decreased interferon signaling and proinflammatory cytokine induction in oHSV-treated tumor cells and inhibited migration of macrophages, resulting in enhanced oHSV replication and cytotoxicity. OS2966 treatment also significantly enhanced oHSV replication and oHSV-mediated antitumor efficacy in orthotopic xenograft models, including triple-negative breast cancer and glioblastoma. The results demonstrated the synergistic potential of the combinatory treatment approach with OS2966 to improve antitumor efficacy of conventional oHSV therapy.


Assuntos
Anticorpos Bloqueadores/uso terapêutico , Herpesvirus Humano 1/fisiologia , Integrina beta1/imunologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Técnicas de Cocultura , Terapia Combinada/métodos , Feminino , Glioma/metabolismo , Glioma/patologia , Glioma/terapia , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Nus , Células RAW 264.7 , Replicação Viral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Sci Rep ; 9(1): 1030, 2019 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-30705293

RESUMO

Monoclonal antibodies targeting cytokines are administered parenterally for the systemic treatment of severe psoriasis. However, systemic exposure to the biologic increases the risk of side-effects including immunosuppression, whereas only a small fraction of the active molecules actually reaches the target organ, the skin. This preclinical study examines the feasibility of delivering a humanized anti-CD29 monoclonal antibody (OS2966) topically to skin using minimally-invasive fractional laser ablation. This approach would enable the targeted use of a biologic for the treatment of recalcitrant psoriatic plaques in patients with less widespread disease while minimizing the risk of systemic exposure. First, the effect of a wide range of laser poration conditions on skin permeation and deposition of OS2966 was tested in vitro to determine optimal microporation parameters. Subsequently, confocal laser scanning microscopy was employed to visualize the distribution of fluorescently-labelled OS2966 in skin. The results demonstrated that delivery of OS2966 into and across skin was feasible. Above fluences of 35.1 J/cm2, skin deposition and permeation were statistically superior to passive delivery reaching values up to 3.7 ± 1.2 µg/cm2 at the most aggressive condition. Selective targeting of the skin was also possible since ≥70% of the OS2966 was delivered locally to the skin. Although nanogramme quantities were able to permeate across skin, these amounts were orders of magnitude lower than levels seen following subcutaneous or intravenous injection and would result in minimal systemic exposure in vivo. The diffusion of fluorescently-labelled OS2966 into the skin surrounding the pores was clearly higher than in intact skin and demonstrated the feasibility of delivering the antibody at least as deep as the dermo-epithelial junction, a critical border region where inflammatory cells cross to promote disease progression. These preliminary results confirm that fractional laser ablation can be used for the cutaneous delivery of OS2966 and now preclinical/clinical studies are required to demonstrate therapeutic efficacy.


Assuntos
Terapia a Laser , Pele/metabolismo , Administração Cutânea , Animais , Anticorpos Monoclonais/imunologia , Ensaio de Imunoadsorção Enzimática , Microscopia Confocal , Absorção Cutânea , Suínos
5.
Cancer Res ; 74(1): 3-7, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24327727

RESUMO

Angiogenesis is an important tissue-level program supporting the growth of highly aggressive cancers and early-stage metastases. However, rapid emergence of resistance to antiangiogenic therapies, such as bevacizumab, greatly limits the clinical utility of these promising approaches. The mechanisms of resistance to antiangiogenic therapy remain incompletely understood. The tumor microenvironment has been demonstrated to be a source of broad therapeutic resistance in multiple cancers. Much of the interaction between the cells comprising a tumor and their microenvironment is driven by integrins. Notably, signaling downstream of integrins in tumor cells promotes fundamental programs vital to aggressive cancer biology, including proliferation, growth, invasion, and survival signaling. These functions then can contribute to malignant phenotypes, including metastasis, therapy resistance, epithelial-to-mesenchymal transition, and angiogenesis. Accordingly, we found ß1 integrin to be functionally upregulated in tumor specimens from patients after bevacizumab failure and in xenograft models of bevacizumab resistance. Inhibition of ß1 in tumor cells with stable gene knockdown or treatment with OS2966, a neutralizing ß1 integrin monoclonal antibody, attenuated aggressive tumor phenotypes in vitro and blocked growth of bevacizumab-resistant tumor xenografts in vivo. Thus, ß1 integrins promote resistance to antiangiogenic therapy through potentiation of multiple malignant programs facilitated by interactions with the tumor microenvironment. The elucidation of this mechanism creates an outstanding opportunity for improving patient outcomes in cancer.


Assuntos
Inibidores da Angiogênese/farmacologia , Integrina beta1/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Técnicas de Silenciamento de Genes , Humanos , Integrina beta1/genética , Neovascularização Patológica/tratamento farmacológico
6.
Cancer Res ; 73(10): 3145-54, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23644530

RESUMO

Antiangiogenic therapies like bevacizumab offer promise for cancer treatment, but acquired resistance, which often includes an aggressive mesenchymal phenotype, can limit the use of these agents. Upregulation of ß1 integrin (ITGB1) occurs in some bevacizumab-resistant glioblastomas (BRG) whereby, mediating tumor-microenvironment interactions, we hypothesized that it may mediate a mesenchymal-type resistance to antiangiogenic therapy. Immunostaining analyses of ß1 integrin and its downstream effector kinase FAK revealed upregulation in 75% and 86% of BRGs, respectively, compared with pretreatment paired specimens. Furthermore, flow cytometry revealed eight-fold more ß1 integrin in primary BRG cells compared with cells from bevacizumab-naïve glioblastomas (BNG). Fluorescence recovery after photobleaching of cells engineered to express a ß1-GFP fusion protein indicated that the mobile ß1 integrin fraction was doubled, and half-life of ß1 integrin turnover in focal adhesions was reduced markedly in BRG cells compared with bevacizumab-responsive glioblastoma multiforme cells. Hypoxia, which was increased with acquisition of bevacizumab resistance, was associated with increased ß1 integrin expression in cultured BNG cells. BRGs displayed an aggressive mesenchymal-like phenotype in vitro. We found that growth of BRG xenograft tumors was attenuated by the ß1 antibody, OS2966, allowing a 20-fold dose reduction of bevacizumab per cycle in this model. Intracranial delivery of OS2966 through osmotic pumps over 28 days increased tumor cell apoptosis, decreased tumor cell invasiveness, and blunted the mesenchymal morphology of tumor cells. We concluded that ß1 integrin upregulation in BRGs likely reflects an onset of hypoxia caused by antiangiogenic therapy, and that ß1 inhibition is well tolerated in vivo as a tractable strategy to disrupt resistance to this therapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Integrina beta1/fisiologia , Animais , Bevacizumab , Neoplasias Encefálicas/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Glioblastoma/patologia , Humanos , Camundongos , Esferoides Celulares/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
7.
Clin Cancer Res ; 19(7): 1773-83, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23307858

RESUMO

PURPOSE: To identify mediators of glioblastoma antiangiogenic therapy resistance and target these mediators in xenografts. EXPERIMENTAL DESIGN: We conducted microarray analysis comparing bevacizumab-resistant glioblastomas (BRG) with pretreatment tumors from the same patients. We established novel xenograft models of antiangiogenic therapy resistance to target candidate resistance mediator(s). RESULTS: BRG microarray analysis revealed upregulation versus pretreatment of receptor tyrosine kinase c-Met, which underwent further investigation because of its prior biologic plausibility as a bevacizumab resistance mediator. BRGs exhibited increased hypoxia versus pretreatment in a manner correlating with their c-Met upregulation, increased c-Met phosphorylation, and increased phosphorylation of c-Met-activated focal adhesion kinase and STAT3. We developed 2 novel xenograft models of antiangiogenic therapy resistance. In the first model, serial bevacizumab treatment of an initially responsive xenograft generated a xenograft with acquired bevacizumab resistance, which exhibited upregulated c-Met expression versus pretreatment. In the second model, a BRG-derived xenograft maintained refractoriness to the MRI tumor vasculature alterations and survival-promoting effects of bevacizumab. Growth of this BRG-derived xenograft was inhibited by a c-Met inhibitor. Transducing these xenograft cells with c-Met short hairpin RNA inhibited their invasion and survival in hypoxia, disrupted their mesenchymal morphology, and converted them from bevacizumab-resistant to bevacizumab-responsive. Engineering bevacizumab-responsive cells to express constitutively active c-Met caused these cells to form bevacizumab-resistant xenografts. CONCLUSION: These findings support the role of c-Met in survival in hypoxia and invasion, features associated with antiangiogenic therapy resistance, and growth and therapeutic resistance of xenografts resistant to antiangiogenic therapy. Therapeutically targeting c-Met could prevent or overcome antiangiogenic therapy resistance.


Assuntos
Inibidores da Angiogênese/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neovascularização Patológica/genética , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Transcriptoma , Inibidores da Angiogênese/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Bevacizumab , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Análise por Conglomerados , Resistencia a Medicamentos Antineoplásicos/genética , Ativação Enzimática/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Humanos , Camundongos , Invasividade Neoplásica/genética , Neovascularização Patológica/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Clin Cancer Res ; 18(10): 2930-42, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22472177

RESUMO

PURPOSE: To identify mechanisms and mediators of resistance to antiangiogenic therapy in human glioblastoma. EXPERIMENTAL DESIGN: We carried out microarray gene expression analysis and immunohistochemistry comparing 21 recurrent glioblastomas progressing during antiangiogenic treatment with VEGF neutralizing antibody bevacizumab to paired pretreatment tumors from the same patients. RESULTS: Microarray analysis revealed that bevacizumab-resistant glioblastomas (BRG) had two clustering patterns defining subtypes that reflect radiographic growth patterns. Enhancing BRGs (EBRG) exhibited MRI enhancement, a long-established criterion for glioblastoma progression, and expressed mitogen-activated protein kinases, neural cell adhesion molecule-1 (NCAM-1), and aquaporin 4. Compared with their paired pretreatment tumors, EBRGs had unchanged vascularity and hypoxia, with increased proliferation. Nonenhancing BRGs (NBRG) exhibited minimal MRI enhancement but had FLAIR-bright expansion, a newer criterion for glioblastoma recurrence since the advent of antiangiogenic therapy, and expressed integrin α5, laminin, fibronectin1, and PDGFRß. NBRGs had less vascularity, more hypoxia, and unchanged proliferation than their paired pretreatment tumors. Primary NBRG cells exhibited more stellate morphology with a 3-fold increased shape factor and were nearly 4-fold more invasive in Matrigel chambers than primary cells from EBRGs or bevacizumab-naive glioblastomas (P < 0.05). CONCLUSION: Using microarray analysis, we found two resistance patterns during antiangiogenic therapy with distinct molecular profiles and radiographic growth patterns. These studies provide valuable biologic insight into the resistance that has limited antiangiogenic therapy to date.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Aquaporina 4/biossíntese , Aquaporina 4/genética , Bevacizumab , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Antígeno CD56/biossíntese , Antígeno CD56/genética , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Progressão da Doença , Fibronectinas/biossíntese , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Integrina alfa5/biossíntese , Laminina/biossíntese , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Proteínas Quinases Ativadas por Mitógeno/genética , Neovascularização Patológica , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/biossíntese , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular
9.
Cancer Res ; 72(7): 1773-83, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22447568

RESUMO

Antiangiogenic therapy leads to devascularization that limits tumor growth. However, the benefits of angiogenesis inhibitors are typically transient and resistance often develops. In this study, we explored the hypothesis that hypoxia caused by antiangiogenic therapy induces tumor cell autophagy as a cytoprotective adaptive response, thereby promoting treatment resistance. Hypoxia-induced autophagy was dependent on signaling through the hypoxia-inducible factor-1α (HIF-1α)/AMPK pathway, and treatment of hypoxic cells with autophagy inhibitors caused a shift from autophagic to apoptotic cell death in vitro. In glioblastomas, clinically resistant to the VEGF-neutralizing antibody bevacizumab, increased regions of hypoxia and higher levels of autophagy-mediating BNIP3 were found when compared with pretreatment specimens from the same patients. When treated with bevacizumab alone, human glioblastoma xenografts showed increased BNIP3 expression and hypoxia-associated growth, which could be prevented by addition of the autophagy inhibitor chloroquine. In vivo targeting of the essential autophagy gene ATG7 also disrupted tumor growth when combined with bevacizumab treatment. Together, our findings elucidate a novel mechanism of resistance to antiangiogenic therapy in which hypoxia-mediated autophagy promotes tumor cell survival. One strong implication of our findings is that autophagy inhibitors may help prevent resistance to antiangiogenic therapy used in the clinic.


Assuntos
Adaptação Fisiológica , Inibidores da Angiogênese/uso terapêutico , Autofagia/fisiologia , Neoplasias Encefálicas/tratamento farmacológico , Hipóxia Celular , Glioblastoma/tratamento farmacológico , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Bevacizumab , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Cloroquina/farmacologia , Glioblastoma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Proteínas de Membrana/análise , Camundongos , Proteínas Proto-Oncogênicas/análise , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Proc Natl Acad Sci U S A ; 109(17): 6674-9, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22451897

RESUMO

Metastasis to the brain is a leading cause of cancer mortality. The current diagnostic method of gadolinium-enhanced MRI is sensitive only to larger tumors, when therapeutic options are limited. Earlier detection of brain metastases is critical for improved treatment. We have developed a targeted MRI contrast agent based on microparticles of iron oxide that enables imaging of endothelial vascular cell adhesion molecule-1 (VCAM-1). Our objectives here were to determine whether VCAM-1 is up-regulated on vessels associated with brain metastases, and if so, whether VCAM-1-targeted MRI enables early detection of these tumors. Early up-regulation of cerebrovascular VCAM-1 expression was evident on tumor-associated vessels in two separate murine models of brain metastasis. Metastases were detectable in vivo using VCAM-1-targeted MRI 5 d after induction (<1,000 cells). At clinical imaging resolutions, this finding is likely to translate to detection at tumor volumes two to three orders of magnitude smaller (0.3-3 × 10(5) cells) than those volumes detectable clinically (10(7)-10(8) cells). VCAM-1 expression detected by MRI increased significantly (P < 0.0001) with tumor progression, and tumors showed no gadolinium enhancement. Importantly, expression of VCAM-1 was shown in human brain tissue containing both established metastases and micrometastases. Translation of this approach to the clinic could increase therapeutic options and change clinical management in a substantial number of cancer patients.


Assuntos
Neoplasias Encefálicas/secundário , Imageamento por Ressonância Magnética/métodos , Animais , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Progressão da Doença , Humanos , Imuno-Histoquímica , Camundongos , Sensibilidade e Especificidade , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/metabolismo
11.
PLoS One ; 4(6): e5857, 2009 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-19516901

RESUMO

Brain-specific homing and direct interactions with the neural substance are prominent hypotheses for brain metastasis formation and a modern manifestation of Paget's "seed and soil" concept. However, there is little direct evidence for this "neurotropic" growth in vivo. In contrast, many experimental studies have anecdotally noted the propensity of metastatic cells to grow along the exterior of pre-existing vessels of the CNS, a process termed vascular cooption. These observations suggest the "soil" for malignant cells in the CNS may well be vascular, rather than neuronal. We used in vivo experimental models of brain metastasis and analysis of human clinical specimens to test this hypothesis. Indeed, over 95% of early micrometastases examined demonstrated vascular cooption with little evidence for isolated neurotropic growth. This vessel interaction was adhesive in nature implicating the vascular basement membrane (VBM) as the active substrate for tumor cell growth in the brain. Accordingly, VBM promoted adhesion and invasion of malignant cells and was sufficient for tumor growth prior to any evidence of angiogenesis. Blockade or loss of the beta1 integrin subunit in tumor cells prevented adhesion to VBM and attenuated metastasis establishment and growth in vivo. Our data establishes a new understanding of CNS metastasis formation and identifies the neurovasculature as the critical partner for such growth. Further, we have elucidated the mechanism of vascular cooption for the first time. These findings may help inform the design of effective molecular therapies for patients with fatal CNS malignancies.


Assuntos
Membrana Basal/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Encéfalo/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/patologia , Humanos , Integrina beta1/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neovascularização Patológica
12.
J Neurosci ; 25(30): 7040-7, 2005 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-16049180

RESUMO

Microglia rapidly become reactive in response to diverse stimuli and are thought to be prominent participants in the pathophysiology of both acute injury and chronic neurological diseases. However, mature microglial reactions to a focal lesion have not been characterized dynamically in adult vertebrate tissue. Here, we present a detailed analysis of long-distance perilesional microglial migration using time-lapse confocal microscopy in acutely isolated living slices from adult brain-injured mice. Extensive migration of perilesional microglia was apparent by 24 h after injury and peaked at 3 d. Average instantaneous migration speeds of approximately 5 microm/min and peak speeds >10 microm/min were observed. Collective, directed migration toward the lesion edge was not observed as might be expected in the presence of chemoattractive gradients. Rather, migration was autonomous and could be modeled as a random walk. Pharmacological blockade of the cysteine-cysteine chemokine receptor 5 reduced migration velocity and the number of perilesional migratory microglia without affecting directional persistence, suggesting a novel role for chemokines in modulation of discrete migratory parameters. Finally, activated microglia in the denervated hippocampal stratum oriens did not migrate extensively, whereas human immunodeficiency virus-1 tat-activated microglia migrated nearly twice as fast as those at the stab lesion, indicating a nonuniform microglial response to different stimuli. Understanding the characteristics and specific molecular mechanisms underlying microglial migration after neural injury could reveal novel targets for therapeutic strategies for modulating neuroinflammation in human diseases.


Assuntos
Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Movimento Celular/imunologia , Microglia/imunologia , Receptores CCR5/metabolismo , Amidas/farmacologia , Animais , Lesões Encefálicas/metabolismo , Antagonistas dos Receptores CCR5 , Movimento Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microscopia Confocal/métodos , Compostos de Amônio Quaternário/farmacologia
13.
J Neuroinflammation ; 2(1): 5, 2005 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-15679892

RESUMO

BACKGROUND: Subventricular microglia (SVMs) are positioned at the interface of the cerebrospinal fluid and brain parenchyma and may play a role in periventricular inflammatory reactions. However, SVMs have not been previously investigated in detail due to the lack of a specific methodology for their study exclusive of deeper parenchymal microglia. METHODS: We have developed and characterized a novel model for the investigation of subventricular microglial reactions in mice using intracerebroventricular (ICV) injection of high-dose rhodamine dyes. Dynamic studies using timelapse confocal microscopy in situ complemented the histopathological analysis. RESULTS: We demonstrate that high-dose ICV rhodamine dye injection resulted in selective uptake by the ependyma and ependymal death within hours. Phagocytosis of ependymal debris by activated SVMs was evident by 1d as demonstrated by the appearance of rhodamine-positive SVMs. In the absence of further manipulation, labelled SVMs remained in the subventricular space. However, these cells exhibited the ability to migrate several hundred microns into the parenchyma towards a deafferentation injury of the hippocampus. This "infiltrative microgliosis" was verified in situ using timelapse confocal microscopy. Finally, supporting the disease relevance of this event, the triad of ependymal cell death, SVM activation, and infiltrative microgliosis was recapitulated by a single ICV injection of HIV-1 tat protein. CONCLUSIONS: Subependymal microglia exhibit robust activation and migration in periventricular inflammatory responses. Further study of this population of microglia may provide insight into neurological diseases with tendencies to involve the ventricular system and periventricular tissues.

14.
J Neurotrauma ; 20(4): 327-36, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12866812

RESUMO

Astrogliosis is a nearly ubiquitous response to a variety of insults to the central nervous system (CNS). This reaction is triggered rapidly, but can persist for years after the initial trauma. Little is known about the signaling mechanisms responsible for this activation and its chronic maintenance. Extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) activation is implicated in several functions important to the reactive glial phenotype such as cellular proliferation and motility. Here we utilize immunohistochemistry with a phosphorylation state-specific antibody (pERK) to characterize the temporal and spatial pattern of ERK/MAPK activation in neurons and glia following a forebrain stab lesion (FSL) in mice. Early activation (1 h) was primarily in perilesional neuronal elements, particularly of the hippocampus. Occasional perilesional glia were also positive for pERK. Additionally, ependymal cells bilaterally stained prominently for pERK. These patterns of pERK immunoreactivity at 1 h were abolished by pretreatment with the selective MEK inhibitor, SL327. ERK/MAPK activation at later time points between 1 day (d) and 30 d was primarily restricted to perilesional astrocytes with maximum labeling at 3 d. However, pERK-positive astrocytes represented only a subset of total GFAP-positive cells and were found more proximal to the lesion suggesting specific functional activation of these cells. Finally, immunostaining for the phosphorylated form of cAMP response element-binding (CREB) protein, a downstream target of the ERK/MAPK cascade, was increased in perilesional glia 7 d after FSL. Sustained activation of the ERK/MAPK signaling pathway in perilesional reactive glia suggests a critical role for this cascade in astrogliosis.


Assuntos
Astrócitos/imunologia , Lesões Encefálicas/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Proteínas Quinases Ativadas por Mitógeno/imunologia , Neurônios/imunologia , Prosencéfalo/imunologia , Prosencéfalo/lesões , Ferimentos Perfurantes/imunologia , Animais , Astrócitos/patologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Prosencéfalo/patologia , Fatores de Tempo , Ferimentos Perfurantes/complicações , Ferimentos Perfurantes/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...