Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(33): e2112006119, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35939683

RESUMO

IL13Rα2 is an attractive target due to its overexpression in a variety of cancers and rare expression in healthy tissue, motivating expansion of interleukin 13 (IL13)-based chimeric antigen receptor (CAR) T cell therapy from glioblastoma into systemic malignancies. IL13Rα1, the other binding partner of IL13, is ubiquitously expressed in healthy tissue, raising concerns about the therapeutic window of systemic administration. IL13 mutants with diminished binding affinity to IL13Rα1 were previously generated by structure-guided protein engineering. In this study, two such variants, termed C4 and D7, are characterized for their ability to mediate IL13Rα2-specific response as binding domains for CAR T cells. Despite IL13Rα1 and IL13Rα2 sharing similar binding interfaces on IL13, mutations to IL13 that decrease binding affinity for IL13Rα1 did not drastically change binding affinity for IL13Rα2. Micromolar affinity to IL13Rα1 was sufficient to pacify IL13-mutein CAR T cells in the presence of IL13Rα1-overexpressing cells in vitro. Interestingly, effector activity of D7 CAR T cells, but not C4 CAR T cells, was demonstrated when cocultured with IL13Rα1/IL4Rα-coexpressing cancer cells. While low-affinity interactions with IL13Rα1 did not result in observable toxicities in mice, in vivo biodistribution studies demonstrated that C4 and D7 CAR T cells were better able to traffic away from IL13Rα1+ lung tissue than were wild-type (WT) CAR T cells. These results demonstrate the utility of structure-guided engineering of ligand-based binding domains with appropriate selectivity while validating IL13-mutein CARs with improved selectivity for application to systemic IL13Rα2-expressing malignancies.


Assuntos
Imunoterapia Adotiva , Subunidade alfa2 de Receptor de Interleucina-13 , Interleucina-13 , Neoplasias , Animais , Linhagem Celular Tumoral , Humanos , Imunoterapia Adotiva/métodos , Interleucina-13/genética , Interleucina-13/farmacocinética , Interleucina-13/uso terapêutico , Subunidade alfa2 de Receptor de Interleucina-13/antagonistas & inibidores , Camundongos , Neoplasias/terapia , Engenharia de Proteínas , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Proc Natl Acad Sci U S A ; 116(3): 997-1006, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30602457

RESUMO

Glioblastoma multiforme (GBM) is a highly aggressive malignant brain tumor with fatal outcome. Tumor-associated macrophages and microglia (TAMs) have been found to be major tumor-promoting immune cells in the tumor microenvironment. Hence, modulation and reeducation of tumor-associated macrophages and microglia in GBM is considered a promising antitumor strategy. Resident microglia and invading macrophages have been shown to have distinct origin and function. Whereas yolk sac-derived microglia reside in the brain, blood-derived monocytes invade the central nervous system only under pathological conditions like tumor formation. We recently showed that disruption of the SIRPα-CD47 signaling axis is efficacious against various brain tumors including GBM primarily by inducing tumor phagocytosis. However, most effects are attributed to macrophages recruited from the periphery but the role of the brain resident microglia is unknown. Here, we sought to utilize a model to distinguish resident microglia and peripheral macrophages within the GBM-TAM pool, using orthotopically xenografted, immunodeficient, and syngeneic mouse models with genetically color-coded macrophages (Ccr2RFP) and microglia (Cx3cr1GFP). We show that even in the absence of phagocytizing macrophages (Ccr2RFP/RFP), microglia are effector cells of tumor cell phagocytosis in response to anti-CD47 blockade. Additionally, macrophages and microglia show distinct morphological and transcriptional changes. Importantly, the transcriptional profile of microglia shows less of an inflammatory response which makes them a promising target for clinical applications.


Assuntos
Neoplasias Encefálicas/imunologia , Antígeno CD47/imunologia , Glioblastoma/imunologia , Microglia/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias Experimentais/imunologia , Fagocitose , Receptores Imunológicos/imunologia , Transdução de Sinais/imunologia , Animais , Neoplasias Encefálicas/patologia , Antígeno CD47/genética , Glioblastoma/genética , Glioblastoma/patologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Microglia/patologia , Monócitos/imunologia , Monócitos/patologia , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Receptores Imunológicos/genética , Transdução de Sinais/genética
3.
J Cell Physiol ; 233(2): 866-879, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28145567

RESUMO

Neuroblastoma (NB) with various clinical presentation is a known childhood malignancy. Despite significant progress in treatment of NB afflicted patients, high risk disease is usually associated with poor outcome, resulting in long-term survival of less that 50%. Known as a disease most commonly originated form the nerve roots, the variants involved in NB imitation and progression remain to be elucidated. The outcome of low to intermediate risk disease is favorable whereas the high risk NB disease with dismal prognosis, positing the necessity of novel approaches for early detection and prognostication of advanced disease. Tailored immunotherapy approaches have shown significant improvement in high-risk NB patients. It has found a link between Gangliosides and progression of NB. The vast majority of neuroblastoma tumors express elevated levels of GD2, opening new insight into using anti-GD2 drugs as potential treatments for NBs. Implication of anti-GD2 monoclonal antibodies for treatment of high risk NBs triggers further investigation to unearth novel biomarkers as prognostic and response biomarker to guide additional multimodal tailored treatment approaches. A growing body of evidence supports the usefulness of miRNAs to evaluate high risk NBs response to anti-GD2 drugs and further prevent drug-related toxicities in refractory or recurrent NBs. miRNAs and circulating proteins in body fluids (plasma and serum) present as potential biomarkers in early detection of NBs. Here, we summarize various biomarkers involved in diagnosis, prognosis and response to treatment in patients with NB. We further attempted to overview prognostic biomarkers in response to treatment with anti-GD2 drugs.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/sangue , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/tratamento farmacológico , Gangliosídeos/antagonistas & inibidores , Imunoterapia/métodos , MicroRNAs/sangue , Neuroblastoma/sangue , Neuroblastoma/tratamento farmacológico , Animais , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Gangliosídeos/imunologia , Humanos , MicroRNAs/genética , Técnicas de Diagnóstico Molecular , Neuroblastoma/genética , Neuroblastoma/imunologia , Valor Preditivo dos Testes , Resultado do Tratamento
4.
BMC Cancer ; 15: 225, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25884624

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most malignant primary brain tumor in adults, with a median survival time of one and a half years. Traditional treatments, including radiation, chemotherapy, and surgery, are not curative, making it imperative to find more effective treatments for this lethal disease. γ-Glutamyl transferase (GGT) is a family of enzymes that was shown to control crucial redox-sensitive functions and to regulate the balance between proliferation and apoptosis. GGT7 is a novel GGT family member that is highly expressed in brain and was previously shown to have decreased expression in gliomas. Since other members of the GGT family were found to be altered in a variety of cancers, we hypothesized that GGT7 could regulate GBM growth and formation. METHODS: To determine if GGT7 is involved in GBM tumorigenesis, we modulated GGT7 expression in two GBM cell lines (U87-MG and U138) and monitored changes in tumorigenicity in vitro and in vivo. RESULTS: We demonstrated for the first time that GBM patients with low GGT7 expression had a worse prognosis and that 87% (7/8) of primary GBM tissue samples showed a 2-fold decrease in GGT7 expression compared to normal brain samples. Exogenous expression of GGT7 resulted in a 2- to 3-fold reduction in proliferation and anchorage-independent growth under minimal growth conditions (1% serum). Decreasing GGT7 expression using either short interfering RNA or short hairpin RNA consistently increased proliferation 1.5- to 2-fold. In addition, intracranial injections of U87-MG cells with reduced GGT7 expression increased tumor growth in mice approximately 2-fold, and decreased mouse survival. To elucidate the mechanism by which GGT7 regulates GBM growth, we analyzed reactive oxygen species (ROS) levels in GBM cells with modulated GGT7 expression. We found that enhanced GGT7 expression reduced ROS levels by 11-33%. CONCLUSION: Our study demonstrates that GGT7 is a novel player in GBM growth and that GGT7 can play a critical role in tumorigenesis by regulating anti-oxidative damage. Loss of GGT7 may increase the cellular ROS levels, inducing GBM occurrence and growth. Our findings suggest that GGT7 can be a promising biomarker and a potential therapeutic target for GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , gama-Glutamiltransferase/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/mortalidade , Glioblastoma/patologia , Xenoenxertos , Humanos , Prognóstico , Espécies Reativas de Oxigênio/metabolismo , gama-Glutamiltransferase/genética
5.
Expert Opin Ther Targets ; 26(11): 949-961, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36527817

RESUMO

INTRODUCTION: Glioblastoma Multiforme (GBM) is one of the fatal cancers of the Central Nervous System (CNS). A variety of reasons exist for why previous immunotherapy strategies, especially Immune Checkpoint Blockers (ICBs), did not work in treating GBM patients. The cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a key immune checkpoint receptor. Its overexpression in cancer and immune cells causes tumor cell progression. CTLA-4 suppresses anti-tumor responses inside the GBM tumor-immune microenvironment. AREAS COVERED: It has been attempted to explain the immunobiology of CTLA-4 as well as its interaction with different immune cells and cancer cells that lead to GBM progression. Additionally, CTLA-4 targeting studies have been reviewed and CTLA-4 combination therapy, as a promising therapeutic target and strategy for GBM immunotherapy, is recommended. EXPERT OPINION: CTLA-4 could be a possible supplement for future cancer immunotherapies of GBM. However, many challenges remain such as the high toxicity of CTLA-4 blockers, and the unresponsiveness of most patients to immunotherapy. For the future clinical success of CTLA-4 blocker therapy, combination approaches with other targeted treatments would be a potentially effective strategy. Going forward, predictive biomarkers can be used to reduce trial timelines and increase the chance of success.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Neoplasias Encefálicas/tratamento farmacológico , Terapia Combinada , Antígeno CTLA-4/uso terapêutico , Glioblastoma/tratamento farmacológico , Imunoterapia , Microambiente Tumoral , Antígenos B7/metabolismo
6.
Cancer Discov ; 11(9): 2248-2265, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33837065

RESUMO

Chimeric antigen receptor (CAR) T cells mediate potent antigen-specific antitumor activity; however, their indirect effects on the endogenous immune system are not well characterized. Remarkably, we demonstrate that CAR T-cell treatment of mouse syngeneic glioblastoma (GBM) activates intratumoral myeloid cells and induces endogenous T-cell memory responses coupled with feed-forward propagation of CAR T-cell responses. IFNγ production by CAR T cells and IFNγ responsiveness of host immune cells are critical for tumor immune landscape remodeling to promote a more activated and less suppressive tumor microenvironment. The clinical relevance of these observations is supported by studies showing that human IL13Rα2-CAR T cells activate patient-derived endogenous T cells and monocytes/macrophages through IFNγ signaling and induce the generation of tumor-specific T-cell responses in a responding patient with GBM. These studies establish that CAR T-cell therapy has the potential to shape the tumor microenvironment, creating a context permissible for eliciting endogenous antitumor immunity. SIGNIFICANCE: Our findings highlight the critical role of IFNγ signaling for a productive CAR T-cell therapy in GBM. We establish that CAR T cells can activate resident myeloid populations and promote endogenous T-cell immunity, emphasizing the importance of host innate and adaptive immunity for CAR T-cell therapy of solid tumors.This article is highlighted in the In This Issue feature, p. 2113.


Assuntos
Glioblastoma/tratamento farmacológico , Imunoterapia Adotiva , Interferon gama/metabolismo , Células Mieloides/imunologia , Receptores de Antígenos Quiméricos/imunologia , Animais , Glioblastoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
JOP ; 11(6): 617-9, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-21068498

RESUMO

CONTEXT: Cystic lymphangiomas originate as benign masses which occur mostly in children especially in the head and neck region and/or the groin. Although abdominal lymphangiomas are rare, they are most commonly reported in adults. In addition, pancreatic involvement is rare. Lymphatic malformation with blockage of the lymphatic flow is the most common etiology leading to the formation of lymphangiomas. Cystic lymphangiomas should always be included in the differential diagnosis of abdominal masses which present with mass effect signs and symptoms. Due to its rarity, it forms a diagnostic and therapeutic challenge for the clinician. CASE REPORT: We herein report the case of a 43-year-old man with a cystic lymphangioma detected in the head of the pancreas and describe the surgical procedure utilized as the therapeutic medium. CONCLUSION: To remove this mass, we utilized a modified approach to a classic pancreaticoduodenectomy. This technique involved resection of the head of the pancreas while preserving the upper 2nd portion of the duodenum and the ampulla of Vater. The result of our 30-month follow-up of this patient has been very satisfactory with no complications.


Assuntos
Linfangioma Cístico/diagnóstico , Linfangioma Cístico/cirurgia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/cirurgia , Adulto , Humanos , Linfangioma Cístico/patologia , Masculino , Modelos Biológicos , Neoplasias Pancreáticas/patologia , Radiografia Abdominal , Tomografia Computadorizada por Raios X
9.
Nat Commun ; 9(1): 4121, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30297829

RESUMO

Medulloblastoma is the most common malignant brain tumor of childhood. Group 3 medulloblastoma, the most aggressive molecular subtype, frequently disseminates through the leptomeningeal cerebral spinal fluid (CSF) spaces in the brain and spinal cord. The mechanism of dissemination through the CSF remains poorly understood, and the molecular pathways involved in medulloblastoma metastasis and self-renewal are largely unknown. Here we show that NOTCH1 signaling pathway regulates both the initiation of metastasis and the self-renewal of medulloblastoma. We identify a mechanism in which NOTCH1 activates BMI1 through the activation of TWIST1. NOTCH1 expression and activity are directly related to medulloblastoma metastasis and decreased survival rate of tumor-bearing mice. Finally, medulloblastoma-bearing mice intrathecally treated with anti-NRR1, a NOTCH1 blocking antibody, present lower frequency of spinal metastasis and higher survival rate. These findings identify NOTCH1 as a pivotal driver of Group 3 medulloblastoma metastasis and self-renewal, supporting the development of therapies targeting this pathway.


Assuntos
Proliferação de Células/genética , Neoplasias Cerebelares/genética , Regulação Neoplásica da Expressão Gênica , Meduloblastoma/genética , Receptor Notch1/genética , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Bloqueadores/farmacologia , Linhagem Celular Tumoral , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/metabolismo , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/metabolismo , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Metástase Neoplásica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Receptor Notch1/imunologia , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
10.
Nat Commun ; 9(1): 4651, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30389946

RESUMO

The original version of this Article omitted Suzana A. Kahn, Siddhartha S. Mitra & Samuel H. Cheshier as jointly supervising authors. This has now been corrected in both the PDF and HTML versions of the Article.

11.
Sci Signal ; 11(547)2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30206138

RESUMO

A major limitation of targeted cancer therapy is the rapid emergence of drug resistance, which often arises through mutations at or downstream of the drug target or through intrinsic resistance of subpopulations of tumor cells. Medulloblastoma (MB), the most common pediatric brain tumor, is no exception, and MBs that are driven by sonic hedgehog (SHH) signaling are particularly aggressive and drug-resistant. To find new drug targets and therapeutics for MB that may be less susceptible to common resistance mechanisms, we used a developmental phosphoproteomics approach in murine granule neuron precursors (GNPs), the developmental cell of origin of MB. The protein kinase CK2 emerged as a driver of hundreds of phosphorylation events during the proliferative, MB-like stage of GNP growth, including the phosphorylation of three of the eight proteins commonly amplified in MB. CK2 was critical to the stabilization and activity of the transcription factor GLI2, a late downstream effector in SHH signaling. CK2 inhibitors decreased the viability of primary SHH-type MB patient cells in culture and blocked the growth of murine MB tumors that were resistant to currently available Hh inhibitors, thereby extending the survival of tumor-bearing mice. Because of structural interactions, one CK2 inhibitor (CX-4945) inhibited both wild-type and mutant CK2, indicating that this drug may avoid at least one common mode of acquired resistance. These findings suggest that CK2 inhibitors may be effective for treating patients with MB and show how phosphoproteomics may be used to gain insight into developmental biology and pathology.


Assuntos
Caseína Quinase II/metabolismo , Neoplasias Cerebelares/metabolismo , Proteínas Hedgehog/metabolismo , Meduloblastoma/metabolismo , Fosfoproteínas/metabolismo , Proteômica/métodos , Transdução de Sinais , Anilidas/farmacologia , Animais , Caseína Quinase II/antagonistas & inibidores , Caseína Quinase II/genética , Linhagem Celular Tumoral , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/genética , Humanos , Estimativa de Kaplan-Meier , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Células NIH 3T3 , Naftiridinas/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Fenazinas , Fosfoproteínas/genética , Piridinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
PLoS One ; 12(9): e0184891, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28934267

RESUMO

Osteosarcoma is the most common primary bone tumor in children and young adults. The median survival of osteosarcoma patients has not significantly improved since 1990, despite administration of different classes of chemotherapy agents, such as methotrexate, cisplatin and doxorubicin. Cancer stem cells (CSCs) are responsible for the resistance of osteosarcoma to chemotherapy and OCT4, SOX2 and SSEA4 have been used to identify CSCs in osteosarcoma. Here, we used low-passage patient-derived osteosarcoma cells and osteosarcoma cells directly isolated from patients before and after chemotherapy treatments to evaluate the effects of chemotherapy on stem cell markers expression. We demonstrate that primary osteosarcoma cells are resistant to methotrexate treatment and sensitive to cisplatin and doxorubicin in vitro. We also verified that cisplatin and doxorubicin reduce the expression of SOX2 and OCT4 in primary osteosarcoma cells whereas methotrexate does not alter SOX2 and OCT4 expression, however it increases SSEA4 expression in primary osteosarcoma cells. Finally, we found that, although the combination treatment cisplatin plus doxorubicin inhibited the in vivo growth of osteosarcoma cells in NOD-SCID gamma mice subcutaneously injected with SaOs2, the combination treatment cisplatin plus doxorubicin plus methotrexate did not inhibit the in vivo growth of these cells. These observations may provide an explanation for the poor response of osteosarcomas to chemotherapy and point to the need of reevaluating the therapeutic strategies for human osteosarcomas.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Metotrexato/uso terapêutico , Osteossarcoma/tratamento farmacológico , Adolescente , Animais , Antimetabólitos Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Neoplasias Ósseas/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Criança , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Humanos , Masculino , Metotrexato/farmacologia , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Transplante de Neoplasias , Osteossarcoma/metabolismo , Adulto Jovem
13.
Sci Transl Med ; 9(381)2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28298418

RESUMO

Morbidity and mortality associated with pediatric malignant primary brain tumors remain high in the absence of effective therapies. Macrophage-mediated phagocytosis of tumor cells via blockade of the anti-phagocytic CD47-SIRPα interaction using anti-CD47 antibodies has shown promise in preclinical xenografts of various human malignancies. We demonstrate the effect of a humanized anti-CD47 antibody, Hu5F9-G4, on five aggressive and etiologically distinct pediatric brain tumors: group 3 medulloblastoma (primary and metastatic), atypical teratoid rhabdoid tumor, primitive neuroectodermal tumor, pediatric glioblastoma, and diffuse intrinsic pontine glioma. Hu5F9-G4 demonstrated therapeutic efficacy in vitro and in vivo in patient-derived orthotopic xenograft models. Intraventricular administration of Hu5F9-G4 further enhanced its activity against disseminated medulloblastoma leptomeningeal disease. Notably, Hu5F9-G4 showed minimal activity against normal human neural cells in vitro and in vivo, a phenomenon reiterated in an immunocompetent allograft glioma model. Thus, Hu5F9-G4 is a potentially safe and effective therapeutic agent for managing multiple pediatric central nervous system malignancies.


Assuntos
Anticorpos/uso terapêutico , Antígenos de Diferenciação/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Antígeno CD47/imunologia , Fagocitose , Receptores Imunológicos/metabolismo , Animais , Anticorpos/farmacologia , Neoplasias Encefálicas/patologia , Proliferação de Células/efeitos dos fármacos , Criança , Modelos Animais de Doenças , Humanos , Imunocompetência , Injeções Intraventriculares , Meduloblastoma/tratamento farmacológico , Meduloblastoma/patologia , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/secundário , Camundongos Endogâmicos C57BL , Modelos Biológicos , Metástase Neoplásica , Fagocitose/efeitos dos fármacos , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nat Med ; 23(4): 450-460, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28288111

RESUMO

Although blood-brain barrier (BBB) compromise is central to the etiology of diverse central nervous system (CNS) disorders, endothelial receptor proteins that control BBB function are poorly defined. The endothelial G-protein-coupled receptor (GPCR) Gpr124 has been reported to be required for normal forebrain angiogenesis and BBB function in mouse embryos, but the role of this receptor in adult animals is unknown. Here Gpr124 conditional knockout (CKO) in the endothelia of adult mice did not affect homeostatic BBB integrity, but resulted in BBB disruption and microvascular hemorrhage in mouse models of both ischemic stroke and glioblastoma, accompanied by reduced cerebrovascular canonical Wnt-ß-catenin signaling. Constitutive activation of Wnt-ß-catenin signaling fully corrected the BBB disruption and hemorrhage defects of Gpr124-CKO mice, with rescue of the endothelial gene tight junction, pericyte coverage and extracellular-matrix deficits. We thus identify Gpr124 as an endothelial GPCR specifically required for endothelial Wnt signaling and BBB integrity under pathological conditions in adult mice. This finding implicates Gpr124 as a potential therapeutic target for human CNS disorders characterized by BBB disruption.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Glioblastoma/genética , Infarto da Artéria Cerebral Média/genética , Hemorragias Intracranianas/genética , Receptores Acoplados a Proteínas G/genética , Junções Íntimas/metabolismo , Animais , Barreira Hematoencefálica/ultraestrutura , Modelos Animais de Doenças , Células Endoteliais/ultraestrutura , Matriz Extracelular/metabolismo , Citometria de Fluxo , Imunofluorescência , Glioblastoma/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Hemorragias Intracranianas/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Microvasos , Pericitos/ultraestrutura , Reação em Cadeia da Polimerase em Tempo Real , Junções Íntimas/ultraestrutura , Via de Sinalização Wnt
15.
Front Surg ; 3: 11, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26973839

RESUMO

Glioblastoma (GBM) is the most devastating brain tumor, with associated poor prognosis. Despite advances in surgery and chemoradiation, the survival of afflicted patients has not improved significantly in the past three decades. Immunotherapy has been heralded as a promising approach in treatment of various cancers; however, the immune privileged environment of the brain usually curbs the optimal expected response in central nervous system malignancies. In addition, GBM cells create an immunosuppressive microenvironment and employ various methods to escape immune surveillance. The purpose of this review is to highlight the strategies by which GBM cells evade the host immune system. Further understanding of these strategies and the biology of this tumor will pave the way for developing novel immunotherapeutic approaches for treatment of GBM.

16.
Int J Dermatol ; 55(6): e332-7, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26910580

RESUMO

BACKGROUND: Altered levels of sex hormones have been observed in many autoimmune disorders, but there is no considerable data about pemphigus. The aim of this study is to compare serum total and free prolactin and dehydroepiandrosterone sulfate (DHEAS) levels between patients with pemphigus and healthy controls and to determine the correlation of these hormones with disease severity. METHODS: This study included 52 newly diagnosed cases of pemphigus and 57 healthy controls. Serum prolactin (total and free) and DHEAS were measured in all subjects. Data analyses were performed using JMP, Version 7. RESULTS: Pemphigus patients had significantly higher levels of total and free serum prolactin (both P = 0.01) and lower levels of DHEAS (P = 0.005) than healthy controls. A significant association was found between severity of pemphigus and total prolactin levels (r = 0.40, P = 0.003). CONCLUSIONS: The patients with pemphigus had higher total and free prolactin and lower DHEAS concentrations, and patients with more severe disease had higher levels of serum total prolactin. These new data may suggest a potential role for sex hormones in the pathogenesis of pemphigus disease and provide new insights for the better management of this chronic and life-threatening disease.


Assuntos
Sulfato de Desidroepiandrosterona/sangue , Pênfigo/sangue , Prolactina/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Adulto Jovem
17.
Eur J Cancer ; 53: 25-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26693896

RESUMO

Melanoma is a life-threatening malignancy with poor prognosis and a relatively high burden of mortality in advanced stages. The efficacy of current available therapeutic strategies is limited, with a survival rate of less than 10%. Despite rapid advances in biomarker-guided drug development in different tumour types, including melanoma, only a very small number of biomarkers have been identified. Recently, microRNAs (miRNAs) have emerged as a molecular regulator in the development and progression of melanoma. Aberrant activation of some known miRNAs, e.g. let-7a and b, miR-148, miR-155, miR-182, miR-200c, miR-211, miR-214, miR-221 and 222, has been recognised to be linked with melanoma-associated genes such as NRAS, microphthalmia-associated transcription factor, receptor tyrosine kinase c-KIT, AP-2 transcription factor, etc. There is accumulating evidence suggesting the potential impact of circulating miRNAs as diagnostic and therapeutic markers in diseases. In addition, miRNAs have turned out to play important roles in drug-resistance mechanisms; suggesting their modulation as a potential approach to overcome chemoresistance. This review highlights recent preclinical and clinical studies on circulating miRNAs and their potential role as diagnosis, and therapeutic targets in melanoma.


Assuntos
Melanoma/diagnóstico , MicroRNAs/metabolismo , Neoplasias Cutâneas/diagnóstico , Biomarcadores Tumorais , Progressão da Doença , Regulação para Baixo/genética , Exossomos/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Melanoma/genética , MicroRNAs/fisiologia , Prognóstico , Neoplasias Cutâneas/genética , Regulação para Cima/genética
18.
Curr Pharm Des ; 22(3): 397-403, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26561061

RESUMO

MicroRNAs (miRNAs) are a class of small regulatory RNAs that control several cellular processes that may contribute to development of cardiovascular disease (CVD) and the pathophysiological consequences of myocardial infarction (MI). Only a very small-numbers of biomarkers in MI (e.g., Troponin) have been identified, which are sufficiently sensitive, specific and robust. There is growing evidence of an association between specific miRNAs in the pathogenesis of MI. miRNAs are transported within the systemic circulation via exosomes and microparticles, and are therefore detectable in blood, urine, saliva, and other fluid compartments. Dysregulation of myocardial-derived miRNAs, such as miR-1, miR-133, miR-499, and miR-208, have been identified as potential biomarkers in MI. Furthermore, alteration of the levels of some miRNAs during stress-induced apoptosis is reported as a novel therapeutic strategy for cardiac disease. Modulation of mir-24 appears to inhibit cardiomyocyte apoptosis, attenuate infarct size, and reduce cardiac dysfunction. A greater knowledge on the molecular mechanism underlying the functional role of emerging miRNAs, could provide novel insights into identifying of new biomarkers. This review highlights several recent preclinical and clinical studies on the role of miRNAs in myocardial infarction; novel miRNA-based therapeutic approaches for therapeutic intervention, and potential circulating miRNA to be served as biomarkers in patients with suspected MI.


Assuntos
MicroRNAs/sangue , MicroRNAs/uso terapêutico , Infarto do Miocárdio/sangue , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/sangue , Diagnóstico Precoce , Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/genética , Terapia de Alvo Molecular , Infarto do Miocárdio/genética , Infarto do Miocárdio/terapia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Troponina T/sangue
19.
PLoS One ; 11(4): e0153550, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27092773

RESUMO

Tumor-associated macrophages (TAMs) represent an important cellular subset within the glioblastoma (WHO grade IV) microenvironment and are a potential therapeutic target. TAMs display a continuum of different polarization states between antitumorigenic M1 and protumorigenic M2 phenotypes, with a lower M1/M2 ratio correlating with worse prognosis. Here, we investigated the effect of macrophage polarization on anti-CD47 antibody-mediated phagocytosis of human glioblastoma cells in vitro, as well as the effect of anti-CD47 on the distribution of M1 versus M2 macrophages within human glioblastoma cells grown in mouse xenografts. Bone marrow-derived mouse macrophages and peripheral blood-derived human macrophages were polarized in vitro toward M1 or M2 phenotypes and verified by flow cytometry. Primary human glioblastoma cell lines were offered as targets to mouse and human M1 or M2 polarized macrophages in vitro. The addition of an anti-CD47 monoclonal antibody led to enhanced tumor-cell phagocytosis by mouse and human M1 and M2 macrophages. In both cases, the anti-CD47-induced phagocytosis by M1 was more prominent than that for M2. Dissected tumors from human glioblastoma xenografted within NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice and treated with anti-CD47 showed a significant increase of M1 macrophages within the tumor. These data show that anti-CD47 treatment leads to enhanced tumor cell phagocytosis by both M1 and M2 macrophage subtypes with a higher phagocytosis rate by M1 macrophages. Furthermore, these data demonstrate that anti-CD47 treatment alone can shift the phenotype of macrophages toward the M1 subtype in vivo.


Assuntos
Anticorpos/farmacologia , Antígeno CD47/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Fagocitose/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Glioblastoma/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Fenótipo
20.
EMBO Mol Med ; 8(5): 511-26, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27138566

RESUMO

A variety of drugs targeting monoamine receptors are routinely used in human pharmacology. We assessed the effect of these drugs on the viability of tumor-initiating cells isolated from patients with glioblastoma. Among the drugs targeting monoamine receptors, we identified prazosin, an α1- and α2B-adrenergic receptor antagonist, as the most potent inducer of patient-derived glioblastoma-initiating cell death. Prazosin triggered apoptosis of glioblastoma-initiating cells and of their differentiated progeny, inhibited glioblastoma growth in orthotopic xenografts of patient-derived glioblastoma-initiating cells, and increased survival of glioblastoma-bearing mice. We found that prazosin acted in glioblastoma-initiating cells independently from adrenergic receptors. Its off-target activity occurred via a PKCδ-dependent inhibition of the AKT pathway, which resulted in caspase-3 activation. Blockade of PKCδ activation prevented all molecular changes observed in prazosin-treated glioblastoma-initiating cells, as well as prazosin-induced apoptosis. Based on these data, we conclude that prazosin, an FDA-approved drug for the control of hypertension, inhibits glioblastoma growth through a PKCδ-dependent mechanism. These findings open up promising prospects for the use of prazosin as an adjuvant therapy for glioblastoma patients.


Assuntos
Antineoplásicos/farmacologia , Reposicionamento de Medicamentos , Glioblastoma/tratamento farmacológico , Proteína Oncogênica v-akt/metabolismo , Prazosina/farmacologia , Proteína Quinase C-delta/metabolismo , Transdução de Sinais , Animais , Anti-Hipertensivos/farmacologia , Apoptose , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Xenoenxertos , Humanos , Camundongos , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA