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










Base de dados
Intervalo de ano de publicação
1.
J Cereb Blood Flow Metab ; : 271678X241261946, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39053491

RESUMO

Stroke is one of the leading causes of death and disability, yet the cellular response to the ischemic insult is poorly understood limiting therapeutic options. Brain pericytes are crucial for maintaining blood-brain barrier (BBB) integrity and are known to be one of the first responders to ischemic stroke. The exact timeline of cellular events after stroke, however, remains elusive. Using the permanent middle cerebral artery occlusion stroke model, we established a detailed timeline of microvascular events after experimental stroke. Our results show that pericytes respond already within 1 hour after the ischemic insult. We find that approximately 30% of the pericyte population dies as early as 1 hour after stroke, while ca 50% express markers that indicate activation. A decrease of endothelial tight junctions, signs of endothelial cell death and reduction in blood vessel length are only detected at time points after the initial pericyte response. Consistently, markers of BBB leakage are observed several hours after pericyte cell death and/or vascular detachment. Our results suggest that the pericyte response to stroke occurs early and precedes both the endothelial response and the BBB breakdown. This highlights pericytes as an important target cell type to develop new diagnostic and therapeutic tools.

2.
Mol Cell Proteomics ; 23(6): 100782, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38705386

RESUMO

Cellular communication within the brain is imperative for maintaining homeostasis and mounting effective responses to pathological triggers like hypoxia. However, a comprehensive understanding of the precise composition and dynamic release of secreted molecules has remained elusive, confined primarily to investigations using isolated monocultures. To overcome these limitations, we utilized the potential of TurboID, a non-toxic biotin ligation enzyme, to capture and enrich secreted proteins specifically originating from human brain pericytes in spheroid cocultures with human endothelial cells and astrocytes. This approach allowed us to characterize the pericyte secretome within a more physiologically relevant multicellular setting encompassing the constituents of the blood-brain barrier. Through a combination of mass spectrometry and multiplex immunoassays, we identified a wide spectrum of different secreted proteins by pericytes. Our findings demonstrate that the pericytes secretome is profoundly shaped by their intercellular communication with other blood-brain barrier-residing cells. Moreover, we identified substantial differences in the secretory profiles between hypoxic and normoxic pericytes. Mass spectrometry analysis showed that hypoxic pericytes in coculture increase their release of signals related to protein secretion, mTOR signaling, and the complement system, while hypoxic pericytes in monocultures showed an upregulation in proliferative pathways including G2M checkpoints, E2F-, and Myc-targets. In addition, hypoxic pericytes show an upregulation of proangiogenic proteins such as VEGFA but display downregulation of canonical proinflammatory cytokines such as CXCL1, MCP-1, and CXCL6. Understanding the specific composition of secreted proteins in the multicellular brain microvasculature is crucial for advancing our knowledge of brain homeostasis and the mechanisms underlying pathology. This study has implications for the identification of targeted therapeutic strategies aimed at modulating microvascular signaling in brain pathologies associated with hypoxia.


Assuntos
Hipóxia Celular , Técnicas de Cocultura , Pericitos , Esferoides Celulares , Pericitos/metabolismo , Humanos , Esferoides Celulares/metabolismo , Secretoma/metabolismo , Células Endoteliais/metabolismo , Astrócitos/metabolismo , Proteômica/métodos , Comunicação Celular , Barreira Hematoencefálica/metabolismo , Células Cultivadas , Encéfalo/metabolismo , Espectrometria de Massas , Transdução de Sinais
3.
Stroke ; 55(3): 558-568, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38323422

RESUMO

BACKGROUND: Blood-based biomarkers have the potential to reflect cerebrovascular signaling after microvascular injury; yet, the detection of cell-specific signaling has proven challenging. Microvesicles retain parental cell surface antigens allowing detection of cell-specific signaling encoded in their cargo. In ischemic stroke, the progression of pathology involves changes in microvascular signaling whereby brain pericytes, perivascular cells wrapping the microcapillaries, are one of the early responders to the ischemic insult. Intercepting the pericyte signaling response peripherally by isolating pericyte-derived microvesicles may provide not only diagnostic information on microvascular injury but also enable monitoring of important pathophysiological mechanisms. METHODS: Plasma samples were collected from patients with acute ischemic stroke (n=39) at 3 time points after stroke onset: 0 to 6 hours, 12 to 24 hours, and 2 to 6 days, and compared with controls (n=39). Pericyte-derived microvesicles were isolated based on cluster of differentiation 140b expression and quantified by flow cytometry. The protein content was evaluated using a proximity extension assay, and vascular signaling pathways were examined using molecular signature hallmarks and gene ontology. RESULTS: In this case-control study, patients with acute ischemic stroke showed significantly increased numbers of pericyte-derived microvesicles (median, stroke versus controls) at 12 to 24 hours (1554 versus 660 microvesicles/µL; P=0.0041) and 2 to 6 days after stroke (1346 versus 660 microvesicles/µL; P=0.0237). Their proteome revealed anti-inflammatory properties mediated via downregulation of Kirsten rat sarcoma virus and IL (interleukin)-6/JAK/STAT3 signaling at 0 to 6 hours, but proangiogenic as well as proinflammatory signals at 12 to 24 hours. Between 2 and 6 days, proteins were mainly associated with vascular remodeling as indicated by activation of Hedgehog signaling in addition to proangiogenic signals. CONCLUSIONS: We demonstrate that the plasma of patients with acute ischemic stroke reflects (1) an early and time-dependent increase of pericyte-derived microvesicles and (2) changes in the protein cargo of microvesicles over time indicating cell signaling specifically related to inflammation and vascular remodeling.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Humanos , AVC Isquêmico/patologia , Pericitos/patologia , Remodelação Vascular , Estudos de Casos e Controles , Proteínas Hedgehog/metabolismo , Encéfalo/patologia , Acidente Vascular Cerebral/patologia , Transdução de Sinais , Biomarcadores/metabolismo
4.
Transl Stroke Res ; 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37378751

RESUMO

The current treatment options for ischemic stroke aim to achieve reperfusion but are time critical. Novel therapeutic approaches that can be given beyond the limited time window of 3-4.5 h are still an unmet need to be addressed to improve stroke outcomes. The lack of oxygen and glucose in the area of ischemic injury initiates a pathological cascade leading to blood-brain barrier (BBB) breakdown, inflammation, and neuronal cell death, a process that may be intercepted to limit stroke progression. Pericytes located at the blood/brain interface are one of the first responders to hypoxia in stroke and therefore a potential target cell for early stroke interventions. Using single-cell RNA sequencing in a mouse model of permanent middle cerebral artery occlusion, we investigated the temporal differences in transcriptomic signatures in pericytes at 1, 12, and 24 h after stroke. Our results reveal a stroke-specific subcluster of pericytes that is present at 12 and 24 h and characterized by the upregulation of genes mainly related to cytokine signaling and immune response. This study identifies temporal transcriptional changes in the acute phase of ischemic stroke that reflect the early response of pericytes to the ischemic insult and its secondary consequences and may constitute potential future therapeutic targets.

5.
Int J Mol Sci ; 24(6)2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36982744

RESUMO

The brain needs sufficient oxygen in order to function normally. This is achieved by a large vascular capillary network ensuring that oxygen supply meets the changing demand of the brain tissue, especially in situations of hypoxia. Brain capillaries are formed by endothelial cells and perivascular pericytes, whereby pericytes in the brain have a particularly high 1:1 ratio to endothelial cells. Pericytes not only have a key location at the blood/brain interface, they also have multiple functions, for example, they maintain blood-brain barrier integrity, play an important role in angiogenesis and have large secretory abilities. This review is specifically focused on both the cellular and the molecular responses of brain pericytes to hypoxia. We discuss the immediate early molecular responses in pericytes, highlighting four transcription factors involved in regulating the majority of transcripts that change between hypoxic and normoxic pericytes and their potential functions. Whilst many hypoxic responses are controlled by hypoxia-inducible factors (HIF), we specifically focus on the role and functional implications of the regulator of G-protein signaling 5 (RGS5) in pericytes, a hypoxia-sensing protein that is regulated independently of HIF. Finally, we describe potential molecular targets of RGS5 in pericytes. These molecular events together contribute to the pericyte response to hypoxia, regulating survival, metabolism, inflammation and induction of angiogenesis.


Assuntos
Células Endoteliais , Pericitos , Humanos , Pericitos/metabolismo , Células Endoteliais/metabolismo , Encéfalo/metabolismo , Hipóxia/metabolismo , Barreira Hematoencefálica/metabolismo , Oxigênio/metabolismo
6.
Biol Open ; 11(10)2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36111549

RESUMO

Adaptive biological mechanisms to hypoxia are crucial to maintain oxygen homeostasis, especially in the brain. Pericytes, cells uniquely positioned at the blood-brain interface, respond fast to hypoxia by expressing regulator of G-protein signalling 5 (RGS5), a negative regulator of G-protein-coupled receptors. RGS5 expression in pericytes is observed in pathological hypoxic environments (e.g. tumours and ischaemic stroke) and associated with perivascular depletion of pericytes and vessel leakage. However, the regulation of RGS5 expression and its functional role in pericytes are not known. We demonstrate that RGS5 acts as a hypoxia-responsive protein in human brain pericytes that is regulated independent of hypoxia inducible factor-1α (HIF-1α), rapidly stabilized under hypoxia, but degraded under normoxic conditions. We show that RGS5 expression desensitizes pericytes to signalling of platelet-derived growth factor-BB (PDGFBB) and sphingosine 1-phosphate (S1P), and blocks chemokinesis or chemotaxis induced by these factors. Our data imply a role for RGS5 in antagonizing pericyte recruitment and retention to blood vessels during hypoxia and support RGS5 as a target in counteracting vessel leakage under pathological hypoxic conditions. This article has an associated First Person interview with the first author of the paper.


Assuntos
Isquemia Encefálica , Pericitos , Proteínas RGS , Acidente Vascular Cerebral , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Proteínas de Ligação ao GTP/metabolismo , Humanos , Hipóxia/metabolismo , Oxigênio , Pericitos/metabolismo , Pericitos/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Acidente Vascular Cerebral/metabolismo
7.
Sci Rep ; 11(1): 1120, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441868

RESUMO

The pathological hallmark of Parkinson's disease (PD) is the formation of Lewy bodies containing aggregated alpha-synuclein (α-syn). Although PD is associated with these distinct histological changes, other pathological features such as microvascular alterations have been linked to neurodegeneration. These changes need to be investigated as they create a hostile brain microenvironment and may contribute to the development and progression of the disease. We use a human α-syn overexpression mouse model that recapitulates some of the pathological features of PD in terms of progressive aggregation of human α-syn, impaired striatal dopamine fiber density, and an age-dependent motor deficit consistent with an impaired dopamine release. We demonstrate for the first time in this model a compromised blood-brain barrier integrity and dynamic changes in vessel morphology from angiogenesis at earlier stages to vascular regression at later stages. The vascular alterations are accompanied by a pathological activation of pericytes already at an early stage without changing overall pericyte density. Our data support and further extend the occurrence of vascular pathology as an important pathophysiological aspect in PD. The model used provides a powerful tool to investigate disease-modifying factors in PD in a temporal sequence that might guide the development of new treatments.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Corpo Estriado/irrigação sanguínea , Modelos Animais de Doenças , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , Pericitos/fisiologia , alfa-Sinucleína/genética , Envelhecimento , Animais , Vasos Sanguíneos/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Células Endoteliais/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora , Neurônios/metabolismo , Neurônios/patologia , Pericitos/patologia , Proteínas Recombinantes de Fusão/metabolismo , Substância Negra/metabolismo , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/metabolismo
8.
Life Sci Alliance ; 3(11)2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32958605

RESUMO

Myeloid-derived suppressor cells (MDSCs) are known to contribute to immune evasion in cancer. However, the function of the human granulocytic (G)-MDSC subset during tumor progression is largely unknown, and there are no established markers for their identification in human tumor specimens. Using gene expression profiling, mass cytometry, and tumor microarrays, we here demonstrate that human G-MDSCs occur as neutrophils at distinct maturation stages, with a disease-specific profile. G-MDSCs derived from patients with metastatic breast cancer and malignant melanoma display a unique immature neutrophil profile, that is more similar to healthy donor neutrophils than to G-MDSCs from sepsis patients. Finally, we show that primary G-MDSCs from metastatic breast cancer patients co-transplanted with breast cancer cells, promote tumor growth, and affect vessel formation, leading to myeloid immune cell exclusion. Our findings reveal a role for human G-MDSC in tumor progression and have clinical implications also for targeted immunotherapy.


Assuntos
Neoplasias da Mama/metabolismo , Células Supressoras Mieloides/metabolismo , Neutrófilos/metabolismo , Adulto , Idoso , Neoplasias da Mama/imunologia , Feminino , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Granulócitos/metabolismo , Granulócitos/fisiologia , Humanos , Imunoterapia/métodos , Melanoma/metabolismo , Pessoa de Meia-Idade , Células Mieloides/metabolismo , Células Supressoras Mieloides/fisiologia , Neutrófilos/fisiologia , Transcriptoma/genética
9.
J Neurosci Res ; 98(5): 826-842, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31758600

RESUMO

Scar formation after injury of the brain or spinal cord is a common event. While glial scar formation by astrocytes has been extensively studied, much less is known about the fibrotic scar, in particular after stroke. Platelet-derived growth factor receptor ß-expressing (PDGFRß+ ) pericytes have been suggested as a source of the fibrotic scar depositing fibrous extracellular matrix (ECM) proteins after detaching from the vessel wall. However, to what extent these parenchymal PDGFRß+ cells contribute to the fibrotic scar and whether targeting these cells affects fibrotic scar formation in stroke is still unclear. Here, we utilize male transgenic mice that after a permanent middle cerebral artery occlusion stroke model have a shift from a parenchymal to a perivascular location of PDGFRß+ cells due to the loss of regulator of G-protein signaling 5 in pericytes. We find that only a small fraction of parenchymal PDGFRß+ cells co-label with type I collagen and fibronectin. Consequently, a reduction in parenchymal PDGFRß+ cells by ca. 50% did not affect the overall type I collagen or fibronectin deposition after stroke. The redistribution of PDGFRß+ cells to a perivascular location, however, resulted in a reduced thickening of the vascular basement membrane and changed the temporal dynamics of glial scar maturation after stroke. We demonstrate that parenchymal PDGFRß+ cells are not the main contributor to the fibrotic ECM, and therefore targeting these cells might not impact on fibrotic scar formation after stroke.


Assuntos
Encéfalo/patologia , Matriz Extracelular/patologia , Gliose/patologia , Pericitos/patologia , Acidente Vascular Cerebral/patologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Fibrose/metabolismo , Fibrose/patologia , Gliose/metabolismo , Masculino , Camundongos , Pericitos/metabolismo , Acidente Vascular Cerebral/metabolismo
10.
PLoS One ; 13(3): e0194146, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29518129

RESUMO

Brain pericytes are important to maintain vascular integrity of the neurovascular unit under both physiological and ischemic conditions. Ischemic stroke is known to induce an inflammatory and hypoxic response due to the lack of oxygen and glucose in the brain tissue. How this early response to ischemia is molecularly regulated in pericytes is largely unknown and may be of importance for future therapeutic targets. Here we evaluate the transcriptional responses in in vitro cultured human brain pericytes after oxygen and/or glucose deprivation. Hypoxia has been widely known to stabilise the transcription factor hypoxia inducible factor 1-alpha (HIF1α) and mediate the induction of hypoxic transcriptional programs after ischemia. However, we find that the transcription factors Jun Proto-Oncogene (c-JUN), Nuclear Factor Of Kappa Light Polypeptide Gene Enhancer In B-Cells (NFκB) and signal transducer and activator of transcription 3 (STAT3) bind genes regulated after 2hours (hs) of omitted glucose and oxygen before HIF1α. Potent HIF1α responses require 6hs of hypoxia to substantiate transcriptional regulation comparable to either c-JUN or STAT3. Phosphorylated STAT3 protein is at its highest after 5 min of oxygen and glucose (OGD) deprivation, whereas maximum HIF1α stabilisation requires 120 min. We show that STAT3 regulates angiogenic and metabolic pathways before HIF1α, suggesting that HIF1α is not the initiating trans-acting factor in the response of pericytes to ischemia.


Assuntos
Encéfalo/metabolismo , Glucose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Oxigênio/metabolismo , Pericitos/metabolismo , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica , Encéfalo/patologia , Hipóxia Celular , Humanos , Pericitos/patologia , Proto-Oncogene Mas
11.
J Pediatr Nurs ; 38: e24-e29, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28939000

RESUMO

PURPOSE: The induction of anesthesia is known to be the most stressful part of the day of surgery for the child. Non-pharmacologic intervention is a field of great interest. The aims of this prospective randomized study were to evaluate if (1). A preoperative visit to the operating theatre would attenuate the anxiety of the child on the day of surgery. (2). A preoperative visit to the operating theatre would attenuate the anxiety of the parent on the day of surgery. DESIGN AND METHODS: Children aged 3-12years and their parents were randomly assigned to the intervention group visiting the operating theatre before surgery and the control group, which never visited there. Anxiety of the children in the preoperative period was measured by using the Swedish version of the modified Yale Preoperative Anxiety Scale (m-YPAS). Parent anxiety was measured by the State-Trait Anxiety Inventory (STAI) instrument. RESULTS: Both the children and their parents showed an increase in anxiety during the day of surgery up to the induction of anesthesia. Children in the intervention group showed no reduction in anxiety compared to the control group. There were no differences in anxiety between the parents in the intervention and the control groups. CONCLUSIONS: Though a preoperative visit to the surgery department and extensive information and therapeutic play does not seem to decrease the anxiety of the children scheduled for surgery and their parents it might be very important as information is highly wanted. Non-pharmacological interventions still need investigation in larger studies.


Assuntos
Ansiedade/prevenção & controle , Educação de Pacientes como Assunto/métodos , Estresse Psicológico/prevenção & controle , Procedimentos Cirúrgicos Operatórios/psicologia , Adaptação Psicológica , Adulto , Fatores Etários , Análise de Variância , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Salas Cirúrgicas , Pais/psicologia , Período Pré-Operatório , Estudos Prospectivos , Valores de Referência , Medição de Risco , Fatores Sexuais , Estatísticas não Paramétricas , Estresse Psicológico/etiologia , Procedimentos Cirúrgicos Operatórios/métodos , Suécia , Resultado do Tratamento
12.
Cell ; 163(2): 324-39, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26451483

RESUMO

Neurodegenerative diseases have been linked to inflammation, but whether altered immunomodulation plays a causative role in neurodegeneration is not clear. We show that lack of cytokine interferon-ß (IFN-ß) signaling causes spontaneous neurodegeneration in the absence of neurodegenerative disease-causing mutant proteins. Mice lacking Ifnb function exhibited motor and cognitive learning impairments with accompanying α-synuclein-containing Lewy bodies in the brain, as well as a reduction in dopaminergic neurons and defective dopamine signaling in the nigrostriatal region. Lack of IFN-ß signaling caused defects in neuronal autophagy prior to α-synucleinopathy, which was associated with accumulation of senescent mitochondria. Recombinant IFN-ß promoted neurite growth and branching, autophagy flux, and α-synuclein degradation in neurons. In addition, lentiviral IFN-ß overexpression prevented dopaminergic neuron loss in a familial Parkinson's disease model. These results indicate a protective role for IFN-ß in neuronal homeostasis and validate Ifnb mutant mice as a model for sporadic Lewy body and Parkinson's disease dementia.


Assuntos
Interferon beta/metabolismo , Neurônios/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Animais , Autofagia , Modelos Animais de Doenças , Terapia Genética , Interferon beta/genética , Interferon beta/uso terapêutico , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Camundongos , Camundongos Endogâmicos C57BL , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Transcriptoma , alfa-Sinucleína/metabolismo
13.
Nat Med ; 20(3): 272-82, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24531377

RESUMO

The defective generation or function of regulatory T (Treg) cells in autoimmune disease contributes to chronic inflammation and tissue injury. We report the identification of FoxA1 as a transcription factor in T cells that, after ectopic expression, confers suppressive properties in a newly identified Treg cell population, herein called FoxA1(+) Treg cells. FoxA1 bound to the Pdl1 promoter, inducing programmed cell death ligand 1 (Pd-l1) expression, which was essential for the FoxA1(+) Treg cells to kill activated T cells. FoxA1(+) Treg cells develop primarily in the central nervous system in response to autoimmune inflammation, have a distinct transcriptional profile and are CD4(+)FoxA1(+)CD47(+)CD69(+)PD-L1(hi)FoxP3(-). Adoptive transfer of stable FoxA1(+) Treg cells inhibited experimental autoimmune encephalomyelitis in a FoxA1-and Pd-l1-dependent manner. The development of FoxA1(+) Treg cells is induced by interferon-ß (IFN-ß) and requires T cell-intrinsic IFN-α/ß receptor (Ifnar) signaling, as the frequency of FoxA1(+) Treg cells was reduced in Ifnb(-/-) and Ifnar(-/-) mice. In individuals with relapsing-remitting multiple sclerosis, clinical response to treatment with IFN-ß was associated with an increased frequency of suppressive FoxA1(+) Treg cells in the blood. These findings suggest that FoxA1 is a lineage-specification factor that is induced by IFN-ß and supports the differentiation and suppressive function of FoxA1(+) Treg cells.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Regulação da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Esclerose Múltipla Recidivante-Remitente/metabolismo , Linfócitos T Reguladores/citologia , Adulto , Animais , Apoptose , Antígeno B7-H1/metabolismo , Diferenciação Celular , Linhagem da Célula , Sistema Nervoso Central/metabolismo , Feminino , Humanos , Terapia de Imunossupressão , Interferon beta/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Fatores de Transcrição/metabolismo
14.
J Neurosci ; 33(35): 14231-45, 2013 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-23986257

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive form of brain tumor. In general, tumor growth requires disruption of the tissue microenvironment, yet how this affects glioma progression is unknown. We studied program death-ligand (PD-L)1 in neurons and gliomas in tumors from GBM patients and associated the findings with clinical outcome. Remarkably, we found that upregulation of PD-L1 by neurons in tumor-adjacent brain tissue (TABT) associated positively with GBM patient survival, whereas lack of neuronal PD-L1 expression was associated with high PD-L1 in tumors and unfavorable prognosis. To understand the molecular mechanism of PD-L1 signaling in neurons, we investigated PD-L1 function in cerebellar and cortical neurons and its impact on gliomas. We discovered that neuronal PD-L1-induced caspase-dependent apoptosis of glioma cells. Because interferon (IFN)-ß induces PD-L1 expression, we studied the functional consequences of neuronal Ifnb gene deletion on PD-L1 signaling and function. Ifnb-/- neurons lacked PD-L1 and were defective in inducing glioma cell death; this effect was reversed on PD-L1 gene transfection. Ifnb-/- mice with intracerebral isografts survived poorly. Similar to the observations in GBM patients, better survival in wild-type mice was associated with high neuronal PD-L1 in TABT and downregulation of PD-L1 in tumors, which was defective in Ifnb-/- mice. Our data indicated that neuronal PD-L1 signaling in brain cells was important for GBM patient survival. Reciprocal PD-L1 regulation in TABT and tumor tissue could be a prognostic biomarker for GBM. Understanding the complex interactions between tumor and adjacent stromal tissue is important in designing targeted GBM therapies.


Assuntos
Antígeno B7-H1/genética , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Neurônios/metabolismo , Adulto , Idoso , Animais , Apoptose , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/diagnóstico , Cerebelo/patologia , Córtex Cerebral/patologia , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Glioblastoma/diagnóstico , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Prognóstico
15.
Cytokine ; 64(1): 112-21, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23972727

RESUMO

Interleukin-4 (IL-4) is a potent antiinflammatory cytokine. However its use in the clinic is hampered by side effects. We here describe the identification of a novel synthetic peptide, termed Ph8, derived from α-helix C of IL-4, which interacts with IL-4 receptor α (IL-4Rα). Employing various cultured genetically engineered cell lines and primary lymphocytes, surface plasmon resonance, qPCR, ELISA and immunoblotting techniques we found that Ph8 bound IL-4Rα and mimicked the anti-inflammatory effects of IL-4 by inhibiting TNF-α production by macrophages in vitro. It induced phosphorylation of STAT6 65kD but inhibited phosphorylation of STAT6 110 kD induced by IL-4 in a B-cell line that expressed the type I receptor. It also inhibited the IL-4-stimulated expression of a STAT6-inducible reporter gene in cells that expressed the type II receptor. Ph8 inhibited the proliferation of Th1/2 cells and downregulated the production of IFN-γ in stimulated Th1 cells. Moreover, Ph8 did not induce any shift in Th1/Th2 profile. This is a favorable effect and it is indicating that Ph8 could block general T cell activation and inflammatory responses without further inducing the side effects generally associated with IL-4 signaling. These data collectively show that Ph8 is only a partial agonist of IL-4 mimicking its desirable properties. In agreement, Ph8 treatment of rats with collagen-induced arthritis, a Th1- and antibody- mediated disease of joint, delayed the manifestation of chronic inflammation and reduced acute inflammation in carrageenan-induced edema. Our findings indicate that Ph8 is a promising potential drug candidate for the treatment of inflammatory diseases.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Subunidade alfa de Receptor de Interleucina-4/metabolismo , Interleucina-4/farmacologia , Fragmentos de Peptídeos/farmacologia , Animais , Artrite Experimental/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Edema/tratamento farmacológico , Células HEK293 , Humanos , Interferon gama/metabolismo , Interleucina-4/análogos & derivados , Interleucina-4/química , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/química , Fosforilação/efeitos dos fármacos , Ligação Proteica , Ratos , Ratos Wistar , Fator de Transcrição STAT6/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
16.
Biochem Biophys Res Commun ; 344(4): 1155-60, 2006 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-16647686

RESUMO

SPI-C is a novel ETS protein that is expressed in B lymphocytes. No target gene for SPI-C has so far been defined. We have performed a yeast two-hybrid screen using SPI-C as bait in order to further analyze the functional role of this orphan transcription factor. We found that SPI-C interacted specifically with the C-terminus of STAT6 in yeast. By co-immunoprecipitation in transfected COS7 cells the physical interaction between SPI-C and STAT6 was confirmed. Furthermore, this protein-protein interaction is functional since we could demonstrate that SPI-C and STAT6 stimulated IL4 induced Iepsilon transcription synergistically but only when both proteins bound to DNA. Thus, a protein interaction between SPI-C and STAT6 is the basis for a novel mechanism for regulation of IL4 induced gene expression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Imunoglobulina E/genética , Fator de Transcrição STAT6/metabolismo , Animais , Sequência de Bases , Células COS , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Células Germinativas/imunologia , Células Germinativas/metabolismo , Humanos , Imunoprecipitação , Interleucina-4/metabolismo , Interleucina-4/farmacologia , Dados de Sequência Molecular , Mapeamento de Interação de Proteínas , Proteínas Proto-Oncogênicas c-ets , Transcrição Gênica/efeitos dos fármacos , Transfecção , Técnicas do Sistema de Duplo-Híbrido
17.
Mol Immunol ; 39(16): 1035-43, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12749910

RESUMO

Mice deficient for SPI-group ETS transcription factors PU.1 or SPI-B fail to generate lymphocytes or do not mount normal antibody mediated immune responses, respectively. PU.1 expression is restricted to B-, T-lymphocytes and macrophages, while SPI-B is expressed in B- and T-lymphocytes. SPI-C is an ETS transcription factor closely related to PU.1 and SPI-B, and expressed temporarily during B-cell development and in macrophages. By deletion and mutation analysis we show that the SPI-C protein has a transactivation domain located to the N-terminus, and that the transactivation activity is reduced to that of the DNA binding domain (DBD) alone when four aspartic acid residues are mutated to alanines. PU.1 and SPI-B regulate transcription from acidic domains located to the N-terminus and by recruiting the co-activator PIP to adjacent sites in a sequence specific manner. In contrast to PU.1 and PIP, SPI-C and PIP were unable to form a distinct ternary complex on the Ig lambda light chain lambda(2-4) enhancer element, suggesting that SPI-C could act both as a positive and negative transcriptional regulator during B-lymphocyte differentiation.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Transativadores/química , Transativadores/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Animais , Ácido Aspártico/genética , Linfócitos B/imunologia , Sítios de Ligação , Células COS , Sequência Consenso , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Fatores Reguladores de Interferon , Macrófagos/imunologia , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Células-Tronco/imunologia , Fatores de Tempo , Transativadores/genética , Fatores de Transcrição/metabolismo
18.
Gene ; 299(1-2): 271-8, 2002 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-12459275

RESUMO

Erythroblast transformation-specific domain (ETS) transcription factors regulate some of the critical molecular mechanisms controlling the differentiation of multipotent haematopoietic progenitor cells into effector B-lymphocytes. The SPI-group ETS-protein transcription factors PU.1 and SPI-B play essential and, although coexpressed and binding to similar DNA sequences, unique roles in B-cell differentiation in mice. Mouse SPI-C is an SPI-group ETS protein expressed temporarily during B-cell development and in macrophages. Here we present the genomic organization of the mouse SPI-C gene, and show by rapid amplification of cDNA ends (5'-RACE) analysis that transcription of the mouse SPI-C mRNA starts at a single site producing a single processed transcript. We have also isolated a cDNA clone encoding the human SPI-C homologue, which displays 65% amino acid identity to the murine protein. In addition, we show that the genomic structure of the human and mouse genes are similar, containing a 5' non-coding exon followed by five coding exons. Human SPI-C mRNA is preferentially detected in foetal and adult spleen, lymph nodes and at lower levels in bone marrow and foetal liver. Finally a phylogenetic prediction analysis of SPI-group protein sequences suggest that the SPI-C proteins form a distinct subgroup, with human SPI-C being closest related to the mouse SPI-C protein.


Assuntos
Proteínas de Ligação a DNA/genética , Perfilação da Expressão Gênica , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Éxons , Genes/genética , Células HeLa , Humanos , Íntrons , Células K562 , Camundongos , Dados de Sequência Molecular , Filogenia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...