Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Front Neurol ; 15: 1378203, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38765267

RESUMO

Introduction: Traumatic brain injury to thalamo-cortical pathways is associated with posttraumatic morbidity. Diffuse mechanical forces to white matter tracts and deep grey matter regions induce an inflammatory response and vascular damage resulting in progressive neurodegeneration. Pro-inflammatory cytokines, including interleukin-1ß (IL-1ß), may contribute to the link between inflammation and the injured capillary network after TBI. This study investigates whether IL-1ß is a key contributor to capillary alterations and changes in pericyte coverage in the thalamus and cortex after TBI. Methods: Animals were subjected to central fluid percussion injury (cFPI), a model of TBI causing widespread axonal and vascular pathology, or sham injury and randomized to receive a neutralizing anti-IL-1ß or a control, anti-cyclosporin A antibody, at 30 min post-injury. Capillary length and pericyte coverage of cortex and thalamus were analyzed by immunohistochemistry at 2- and 7-days post-injury. Results and Conclusion: Our results show that early post-injury attenuation of IL-1ß dependent inflammatory signaling prevents capillary damage by increasing pericyte coverage in the thalamus.

2.
Leuk Res ; 140: 107484, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38520796

RESUMO

INTRODUCTION: Thrombocytopenia is among the most common complications following hematopoietic stem cell transplantation and is associated with increased mortality and morbidity with no standard treatment yet. In this multicenter and retrospective study, we aim to present our multi-center experience of Eltrombopag treatment in patients with isolated thrombocytopenia following HSCT. MATERIAL-METHOD: A total of 73 patients from 5 centers who underwent autologous or allogeneic stem cell transplantation, had no primary disease relapse, all of whom had neutrophil engraftment, complete chimerism, and who were diagnosed with Prolonged Isolated Thrombocytopenia (PIT) or Secondary Failure Of Platelet Recovery (SFPR) were included in the study. The patients were initiated on Eltrombopag at a dose of 50-150 mg. Complete response was defined as a platelet count >50×109/L for 7 consecutive days with no transfusion support. RESULTS: A total of 50.3% of the patients underwent Autologous and 49.7% Allogeneic Stem Cell Transplantation, 54.8% were diagnosed with PIT, and 45.2% were diagnosed with SFPR, and the treatment with 50-150 mg/day Eltrombopag was initiated on the median day +42. Complete response was achieved in 71.2% of these patients on the median day 23 of the treatment. No significant effects of the initial dose (50-150 mg/day) were detected in the Complete Response in the multivariate analysis on response. An insufficient number of Megakaryocytes in the bone marrow before Eltrombopag treatment was determined as an independent risk factor in determining the response (OR 3.57, 95% CI 1.21-10.55). The overall survival of the patients who did not respond to Eltrombopag was found to be significantly worse than that of patients who responded (p=0.022, HR:2.74, 95% CI 1.12-6.54). CONCLUSION: As a result of the present study, Eltrombopag treatment was found to be effective and safe in thrombocytopenia that develops following hematopoietic stem cell transplantation. It was concluded that its use may be more effective in patients with sufficient bone marrow megakaryocytes before the treatment and an initial dose of 50 mg/day may be appropriate in terms of cost, effectiveness, and toxicity. Large-scale randomized and controlled prospective studies are needed to determine the roles of Eltrombopag treatment in patients with post-transplant PIT and SFPR.


Assuntos
Benzoatos , Transplante de Células-Tronco Hematopoéticas , Hidrazinas , Pirazóis , Trombocitopenia , Humanos , Hidrazinas/uso terapêutico , Hidrazinas/administração & dosagem , Hidrazinas/efeitos adversos , Benzoatos/uso terapêutico , Benzoatos/administração & dosagem , Benzoatos/efeitos adversos , Pirazóis/uso terapêutico , Pirazóis/efeitos adversos , Pirazóis/administração & dosagem , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Feminino , Masculino , Trombocitopenia/etiologia , Trombocitopenia/tratamento farmacológico , Adulto , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem , Adolescente , Idoso , Contagem de Plaquetas
3.
Glia ; 72(4): 728-747, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38180164

RESUMO

Senescence is a negative prognostic factor for outcome and recovery following traumatic brain injury (TBI). TBI-induced white matter injury may be partially due to oligodendrocyte demise. We hypothesized that the regenerative capacity of oligodendrocyte precursor cells (OPCs) declines with age. To test this hypothesis, the regenerative capability of OPCs in young [(10 weeks ±2 (SD)] and aged [(62 weeks ±10 (SD)] mice was studied in mice subjected to central fluid percussion injury (cFPI), a TBI model causing widespread white matter injury. Proliferating OPCs were assessed by immunohistochemistry for the proliferating cell nuclear antigen (PCNA) marker and labeled by 5-ethynyl-2'-deoxyuridine (EdU) administered daily through intraperitoneal injections (50 mg/kg) from day 2 to day 6 after cFPI. Proliferating OPCs were quantified in the corpus callosum and external capsule on day 2 and 7 post-injury (dpi). The number of PCNA/Olig2-positive and EdU/Olig2-positive cells were increased at 2dpi (p < .01) and 7dpi (p < .01), respectively, in young mice subjected to cFPI, changes not observed in aged mice. Proliferating Olig2+/Nestin+ cells were less common (p < .05) in the white matter of brain-injured aged mice, without difference in proliferating Olig2+/PDGFRα+ cells, indicating a diminished proliferation of progenitors with different spatial origin. Following TBI, co-staining for EdU/CC1/Olig2 revealed a reduced number of newly generated mature oligodendrocytes in the white matter of aged mice when compared to the young, brain-injured mice (p < .05). We observed an age-related decline of oligodendrogenesis following experimental TBI that may contribute to the worse outcome of elderly patients following TBI.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Substância Branca , Humanos , Idoso , Camundongos , Animais , Antígeno Nuclear de Célula em Proliferação , Encéfalo , Oligodendroglia , Camundongos Endogâmicos C57BL
4.
Acta Neuropathol Commun ; 10(1): 129, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-36064443

RESUMO

Cerebellar dysfunction is commonly observed following traumatic brain injury (TBI). While direct impact to the cerebellum by TBI is rare, cerebellar pathology may be caused by indirect injury via cortico-cerebellar pathways. To address the hypothesis that degeneration of Purkinje cells (PCs), which constitute the sole output from the cerebellum, is linked to long-range axonal injury and demyelination, we used the central fluid percussion injury (cFPI) model of widespread traumatic axonal injury in mice. Compared to controls, TBI resulted in early PC loss accompanied by alterations in the size of pinceau synapses and levels of non-phosphorylated neurofilament in PCs. A combination of vDISCO tissue clearing technique and immunohistochemistry for vesicular glutamate transporter type 2 show that diffuse TBI decreased mossy and climbing fiber synapses on PCs. At 2 days post-injury, numerous axonal varicosities were found in the cerebellum supported by fractional anisotropy measurements using 9.4 T MRI. The disruption and demyelination of the cortico-cerebellar circuits was associated with poor performance of brain-injured mice in the beam-walk test. Despite a lack of direct input from the injury site to the cerebellum, these findings argue for novel long-range mechanisms causing Purkinje cell injury that likely contribute to cerebellar dysfunction after TBI.


Assuntos
Lesões Encefálicas Traumáticas , Doenças Desmielinizantes , Animais , Axônios/patologia , Lesões Encefálicas Traumáticas/patologia , Cerebelo/patologia , Doenças Desmielinizantes/patologia , Camundongos , Células de Purkinje/patologia
5.
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.
J Neurotrauma ; 39(5-6): 411-422, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35018831

RESUMO

Cerebellar dysfunction after traumatic brain injury (TBI) is commonly suspected based on clinical symptoms, although cerebellar pathology has rarely been investigated. To address the hypothesis that the cerebellar axon-myelin unit is altered by diffuse TBI, we used the central fluid percussion injury (cFPI) model in adult mice to create widespread axonal injury by delivering the impact to the forebrain. We specifically focused on changes in myelin components (myelin basic protein [MBP], 2',3'-cyclic nucleotide 3'-phosphodiesterase [CNPase], nodal/paranodal domains [neurofascin (Nfasc), ankyrin-G], and phosphorylated neurofilaments [SMI-31, SMI-312]) in the cerebellum, remote from the impact, at two, seven, and 30 days post-injury (dpi). When compared with sham-injured controls, cerebellar MBP and CNPase protein levels were decreased at 2 dpi that remained reduced up to 30 dpi. Diffuse TBI induced different effects on neuronal (Nfasc 186, Nfasc 140) and glial (Nfasc 155) neurofascin isoforms that play a key role in the assembly of the nodes of Ranvier. Expression of Nfasc 140 in the cerebellum increased at 7 dpi, in contrast to Nfasc 155 levels, which were decreased. Although neurofascin binding partner ankyrin-G protein levels decreased acutely after cFPI, its expression levels increased at 7 dpi and remained unchanged up to 30 dpi. The TBI-induced reduction in neurofilament phosphorylation (SMI-31) observed in the cerebellum was closely associated with decreased levels of the myelin proteins MBP and CNPase. This is the first evidence of temporal and spatial structural changes in the axon-myelin unit in the cerebellum, remote from the location of the impact site, in a diffuse TBI model in mice.


Assuntos
Lesões Encefálicas Difusas , Lesões Encefálicas Traumáticas , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Animais , Anquirinas/metabolismo , Axônios/patologia , Lesões Encefálicas Difusas/patologia , Lesões Encefálicas Traumáticas/patologia , Cerebelo/metabolismo , Camundongos , Bainha de Mielina/metabolismo
8.
Cell Death Dis ; 11(2): 98, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32029735

RESUMO

Amyloid beta peptides (Aß) proteins play a key role in vascular pathology in Alzheimer's Disease (AD) including impairment of the blood-brain barrier and aberrant angiogenesis. Although previous work has demonstrated a pro-angiogenic role of Aß, the exact mechanisms by which amyloid precursor protein (APP) processing and endothelial angiogenic signalling cascades interact in AD remain a largely unsolved problem. Here, we report that increased endothelial sprouting in human-APP transgenic mouse (TgCRND8) tissue is dependent on ß-secretase (BACE1) processing of APP. Higher levels of Aß processing in TgCRND8 tissue coincides with decreased NOTCH3/JAG1 signalling, overproduction of endothelial filopodia and increased numbers of vascular pericytes. Using a novel in vitro approach to study sprouting angiogenesis in TgCRND8 organotypic brain slice cultures (OBSCs), we find that BACE1 inhibition normalises excessive endothelial filopodia formation and restores NOTCH3 signalling. These data present the first evidence for the potential of BACE1 inhibition as an effective therapeutic target for aberrant angiogenesis in AD.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Córtex Cerebral/irrigação sanguínea , Células Endoteliais/enzimologia , Neovascularização Patológica , Receptor Notch3/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Inibidores Enzimáticos/farmacologia , Feminino , Técnicas In Vitro , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Densidade Microvascular , Pseudópodes/enzimologia , Pseudópodes/patologia , Transdução de Sinais
9.
Int J Mol Sci ; 21(2)2020 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-31936248

RESUMO

Traumatic brain injury (TBI) increases the risk of delayed neurodegenerative processes, including Parkinson's disease (PD). Interleukin-1beta (IL-1ß), a key pro-inflammatory cytokine, may promote secondary injury development after TBI. Conversely, neutralizing IL-1ß was found to improve functional recovery following experimental TBI. However, the mechanisms underlying the behavioral improvements observed by IL-1ß neutralization are still poorly understood. The present study investigated the role of IL-1ß on the microglia response and neuronal changes in the globus pallidus in response to diffuse TBI. Mice were subjected to sham injury or the central fluid percussion injury (cFPI) (a model of traumatic axonal injury), and were randomly administered an IL-1ß neutralizing or a control antibody at 30 min post-injury. The animals were analyzed at 2, 7, or 14 days post-injury. When compared to controls, mice subjected to cFPI TBI had increased microglia activation and dopaminergic innervation in the globus pallidus, and a decreased number of parvalbumin (PV) positive interneurons in the globus pallidus. Neutralization of IL-1ß attenuated the microglia activation, prevented the loss of PV+ interneurons and normalized dopaminergic fiber density in the globus pallidus of brain-injured animals. These findings argue for an important role for neuro-inflammation in the PD-like pathology observed in TBI.


Assuntos
Anticorpos Neutralizantes/farmacologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Interleucina-1beta/farmacologia , Doença de Parkinson/tratamento farmacológico , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Comportamento Animal/efeitos dos fármacos , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/patologia , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Globo Pálido/efeitos dos fármacos , Globo Pálido/patologia , Humanos , Interleucina-1beta/genética , Ativação de Macrófagos/efeitos dos fármacos , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia
10.
FASEB J ; 33(8): 8990-8998, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31039042

RESUMO

Poststroke recovery requires multiple repair mechanisms, including vascular remodeling and blood-brain barrier (BBB) restoration. Brain pericytes are essential for BBB repair and angiogenesis after stroke, but they also give rise to scar-forming platelet-derived growth factor receptor ß (PDGFR-ß)-expressing cells. However, many of the molecular mechanisms underlying this pericyte response after stroke still remain unknown. Regulator of G-protein signaling 5 (RGS5) has been associated with pericyte detachment from the vascular wall, but whether it regulates pericyte function and vascular stabilization in the chronic phase of stroke is not known. Using RGS5-knockout (KO) mice, we study how loss of RGS5 affects the pericyte response and vascular remodeling in a stroke model at 7 d after ischemia. Loss of RGS5 leads to a shift toward an increase in the number of perivascular pericytes and reduction in the density of parenchymal PDGFR-ß-expressing cells associated with normalized PDGFR-ß activation after stroke. The redistribution of pericytes resulted in higher pericyte coverage, increased vascular density, preservation of vessel lengths, and a significant reduction in vascular leakage in RGS5-KO mice compared with controls. Our study demonstrates RGS5 in pericytes as an important target to enhance vascular remodeling.-Roth, M., Gaceb, A., Enström, A., Padel, T., Genové, G., Özen, I., Paul, G. Regulator of G-protein signaling 5 regulates the shift from perivascular to parenchymal pericytes in the chronic phase after stroke.


Assuntos
Pericitos/metabolismo , Proteínas RGS/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Barreira Hematoencefálica , Capilares/metabolismo , Capilares/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica , Pericitos/patologia , Proteínas RGS/deficiência , Proteínas RGS/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/patologia , Fatores de Tempo
11.
Stroke ; 49(9): 2182-2190, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30354999

RESUMO

Background and Purpose- In ischemic stroke, breakdown of the blood-brain barrier (BBB) aggravates brain damage. Pericyte detachment contributes to BBB disruption and neurovascular dysfunction, but little is known about its regulation in stroke. Here, we investigated how loss of RGS5 (regulator of G protein signaling 5) in pericytes affects BBB breakdown in stroke and its consequences. Method- We used RGS5 knockout and control mice and applied a permanent middle cerebral occlusion model. We analyzed pericyte numbers, phenotype, and vessel morphology using immunohistochemistry and confocal microscopy. We investigated BBB breakdown by measuring endothelial coverage, tight junctions, and AQP4 (aquaporin 4) in addition to BBB permeability (fluorescent-conjugated dextran extravasation). Tissue hypoxia was assessed with pimonidazole hydrochloride and neuronal death quantified with the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Results- We demonstrate that loss of RGS5 increases pericyte numbers and their endothelial coverage, which is associated with higher capillary density and length, and significantly less BBB damage after stroke. Loss of RGS5 in pericytes results in reduced vascular leakage and preserved tight junctions and AQP4, decreased cerebral hypoxia, and partial neuronal protection in the infarct area. Conclusions- Our findings show that loss of RGS5 affects pericyte-related BBB preservation in stroke and identifies RGS5 as an important target for neurovascular protection.


Assuntos
Barreira Hematoencefálica/metabolismo , Endotélio Vascular/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Neurônios/metabolismo , Pericitos/patologia , Proteínas RGS/genética , Junções Íntimas/metabolismo , Animais , Aquaporina 4/metabolismo , Barreira Hematoencefálica/patologia , Permeabilidade Capilar , Morte Celular , Modelos Animais de Doenças , Endotélio Vascular/patologia , Hipóxia/metabolismo , Hipóxia/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média/patologia , Camundongos Knockout , Microscopia Confocal , Neurônios/patologia , Acidente Vascular Cerebral , Junções Íntimas/patologia
12.
Biochimie ; 155: 16-25, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29698670

RESUMO

Personalized and regenerative medicine is an emerging therapeutic strategy that is based on cell biology and biomedical engineering used to develop biological substitutes to maintain normal function or restore damaged tissues and organs. The secretory capacities of different cell types are now explored as such possible therapeutic regenerative agents in a variety of diseases. A secretome can comprise chemokines, cytokines, growth factors, but also extracellular matrix components, microvesicles and exosomes as well as genetic material and may differ depending on the tissue and the stimulus applied to the cell. With regard to clinical applications, the secretome of mesenchymal stem cells (MSC) is currently the most widely explored. However, other cell types such as pericytes may have similar properties as MSC and the potential therapeutic possibilities of these cells are only just beginning to emerge. In this review, we will summarize the currently available data describing the secretome of pericytes and its potential implications for tissue regeneration, whereby we especially focus on brain pericytes as potential new target cell for neuroregeneration and brain repair.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Citocinas/metabolismo , Pericitos/metabolismo , Proteoma/metabolismo , Regeneração , Animais , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Pericitos/citologia , Medicina Regenerativa/métodos
13.
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
14.
J Cereb Blood Flow Metab ; 38(1): 45-57, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28741407

RESUMO

Brain pericytes not only maintain the anatomical, biochemical and immune blood-brain barrier, but display features of mesenchymal stem cells (MSCs) in vitro. MSCs have pro-regenerative properties attributed to their secretome. However, whether also brain pericytes possess such pro-regenerative capacities is largely unknown. Here we characterize the secretome and microvesicle (MV) release of human brain pericytes mediated by platelet-derived growth factor-BB (PDGF-BB)/PDGF receptor beta (PDGFRß) signalling. Upon PDGF-BB, pericytes release not only a plethora of growth factors and a panel of cytokines, but also MVs containing BDNF, FGFb, ßNGF, VEGF and PLGF, a response that is specific for PDGFRß signalling and activation of the ERK 1/2 pathway. In contrast, lipopolysaccharide (LPS), an activator of the innate immune system, stimulates the secretion of much higher amounts of mainly inflammatory cytokines and activates the NFκB pathway. Pericytes change their morphology and undergo opposite changes in surface marker expression, respectively. Our findings provide evidence that the secretome of human brain pericytes varies greatly depending on the exogenous stimulus. The differential secretory functions of pericytes may play an important role in either regulating neuroinflammation or contributing to neurorestoration and identify a possible new target cell for neuroregeneration.


Assuntos
Becaplermina/metabolismo , Encéfalo/metabolismo , Pericitos/metabolismo , Becaplermina/farmacologia , Micropartículas Derivadas de Células/metabolismo , Citocinas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pericitos/efeitos dos fármacos
15.
Neurobiol Dis ; 94: 95-105, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27288154

RESUMO

Parkinson's disease (PD) is a neurodegenerative disease where the degeneration of the nigrostriatal pathway leads to specific motor deficits. There is an unmet medical need for regenerative treatments that stop or reverse disease progression. Several growth factors have been investigated in clinical trials to restore the dopaminergic nigrostriatal pathway damaged in PD. Platelet-derived growth factor-BB (PDGF-BB), a molecule that recruits pericytes to stabilize microvessels, was recently investigated in a phase-1 clinical trial, showing a dose-dependent increase in dopamine transporter binding in the putamen of PD patients. Interestingly, evidence is accumulating that PD is paralleled by microvascular changes, however, whether PDGF-BB modifies pericytes in PD is not known. Using a pericyte reporter mouse strain, we investigate the functional and restorative effect of PDGF-BB in a partial 6-hydroxydopamine medial forebrain bundle lesion mouse model of PD, and whether this restorative effect is accompanied by changes in pericyte features. We demonstrate that a 2-week treatment with PDGF-BB leads to behavioural recovery using several behavioural tests, and partially restores the nigrostriatal pathway. Interestingly, we find that pericytes are activated in the striatum of PD lesioned mice and that these changes are reversed by PDGF-BB treatment. The modulation of brain pericytes may contribute to the PDGF-BB-induced neurorestorative effects, PDGF-BB allowing for vascular stabilization in PD. Pericytes might be a new cell target of interest for future regenerative therapies.


Assuntos
Atividade Motora/efeitos dos fármacos , Doença de Parkinson/metabolismo , Pericitos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/farmacologia , Animais , Becaplermina , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Masculino , Feixe Prosencefálico Mediano/efeitos dos fármacos , Feixe Prosencefálico Mediano/metabolismo , Camundongos Transgênicos , Atividade Motora/fisiologia , Oxidopamina/farmacologia , Doença de Parkinson/patologia , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo
16.
PLoS One ; 10(4): e0123553, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25875288

RESUMO

Glioblastoma multiforme (GBM) is the most common brain tumor in adults. It presents an extremely challenging clinical problem, and treatment very frequently fails due to the infiltrative growth, facilitated by extensive angiogenesis and neovascularization. Pericytes constitute an important part of the GBM microvasculature. The contribution of endogenous brain pericytes to the tumor vasculature in GBM is, however, unclear. In this study, we determine the site of activation and the extent of contribution of endogenous brain pericytes to the GBM vasculature. GL261 mouse glioma was orthotopically implanted in mice expressing green fluorescent protein (GFP) under the pericyte marker regulator of G protein signaling 5 (RGS5). Host pericytes were not only activated within the glioma, but also in cortical areas overlying the tumor, the ipsilateral subventricular zone and within the hemisphere contralateral to the tumor. The host-derived activated pericytes that infiltrated the glioma were mainly localized to the tumor vessel wall. Infiltrating GFP positive pericytes co-expressed the pericyte markers platelet-derived growth factor receptor-ß (PDGFR-ß) and neuron-glial antigen 2. Interestingly, more than half of all PDGFR-ß positive pericytes within the tumor were contributed by the host brain. We did not find any evidence that RGS5 positive pericytes adopt another phenotype within glioma in this paradigm. We conclude that endogenous pericytes become activated in widespread areas of the brain in response to an orthotopic mouse glioma. Host pericytes are recruited into the tumor and constitute a major part of the tumor pericyte population.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Encéfalo/patologia , Glioma/irrigação sanguínea , Microvasos/patologia , Neovascularização Patológica/patologia , Pericitos/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/patologia , Adesão Celular , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Glioma/patologia , Proteínas de Fluorescência Verde/metabolismo , Inflamação/patologia , Laminina/metabolismo , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia
17.
Acta Neuropathol ; 128(3): 381-96, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24848101

RESUMO

Pericytes are located on the abluminal side of endothelial cells lining the microvasculature in all organs. They have been identified as multipotent progenitor cells in several tissues of the body including the human brain. New evidence suggests that pericytes contribute to tissue repair, but their role in the injured brain is largely unknown. Here, we investigate the role of pericytes in ischemic stroke. Using a pericyte-reporter mouse model, we provide unique evidence that regulator of G-protein signaling 5 expressing cells are activated pericytes that leave the blood vessel wall, proliferate and give rise to microglial cells after ischemic brain injury. Consistently, we show that activated pericytes express microglial markers in human stroke brain tissue. We demonstrate that human brain-derived pericytes adopt a microglial phenotype and upregulate mRNA specific for activated microglial cells under hypoxic conditions in vitro. Our study indicates that the vasculature is a novel source of inflammatory cells with a microglial phenotype in brain ischemia and hence identifies pericytes as an important new target for the development of future stroke therapies.


Assuntos
Encéfalo/patologia , Microglia/patologia , Pericitos/patologia , Acidente Vascular Cerebral/patologia , Animais , Antígenos CD/metabolismo , Proteínas de Arabidopsis/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Glucose/deficiência , Humanos , Transferases Intramoleculares/metabolismo , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Microglia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Fatores de Tempo
18.
PLoS One ; 7(4): e35577, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22523602

RESUMO

Blood vessels and adjacent cells form perivascular stem cell niches in adult tissues. In this perivascular niche, a stem cell with mesenchymal characteristics was recently identified in some adult somatic tissues. These cells are pericytes that line the microvasculature, express mesenchymal markers and differentiate into mesodermal lineages but might even have the capacity to generate tissue-specific cell types. Here, we isolated, purified and characterized a previously unrecognized progenitor population from two different regions in the adult human brain, the ventricular wall and the neocortex. We show that these cells co-express markers for mesenchymal stem cells and pericytes in vivo and in vitro, but do not express glial, neuronal progenitor, hematopoietic, endothelial or microglial markers in their native state. Furthermore, we demonstrate at a clonal level that these progenitors have true multilineage potential towards both, the mesodermal and neuroectodermal phenotype. They can be epigenetically induced in vitro into adipocytes, chondroblasts and osteoblasts but also into glial cells and immature neurons. This progenitor population exhibits long-term proliferation, karyotype stability and retention of phenotype and multipotency following extensive propagation. Thus, we provide evidence that the vascular niche in the adult human brain harbors a novel progenitor with multilineage capacity that appears to represent mesenchymal stem cells and is different from any previously described human neural stem cell. Future studies will elucidate whether these cells may play a role for disease or may represent a reservoir that can be exploited in efforts to repair the diseased human brain.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Multipotentes/citologia , Actinas/biossíntese , Adulto , Diferenciação Celular , Linhagem da Célula , Ventrículos Cerebrais/citologia , Células Clonais , Humanos , Neocórtex/citologia , Pericitos/citologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/biossíntese
19.
Clin Transl Med ; 1(1): 30, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23369339

RESUMO

Perivascular adult stem cells have been isolated from several tissues, including the adult human brain. They have unique signatures resembling both pericytes and mesenchymal stem cells. Understanding the nature of these cells in their specific vascular niches is important to determine their clinical potential as a new adult stem cell source. Indeed, they have promising features in vitro in terms of multipotency, immunomodulation and secretion of growth factors and cytokines. However, their in vivo function is less known as yet. Recent emerging data show a crucial role of perivascular mesenchymal stem cells in tissue homeostasis and repair. Furthermore, these cells may play an important role in adult stem cell niche regulation and in neurodegeneration. Here we review the recent literature on perivascular mesenchymal stem cells, discuss their different in vitro functions and highlight especially the specific properties of brain-derived perivascular mesenchymal stem cells. We summarize current evidence that suggests an important in vivo function of these cells in terms of their regenerative potential that may indicate a new target cell for endogenous tissue regeneration and repair.

20.
Brain Inj ; 25(1): 113-26, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21117911

RESUMO

PRIMARY OBJECTIVE: The present study was undertaken to evaluate whether enoant, which is rich in polyphenols, has any effect on electroencephalogram (EEG), oxidative stress and inflammation in ischemia/reperfusion (I/R) injury. METHODS: Ischemia was induced by 2-hour occlusion of bilateral common carotid artery. Animals orally received enoant. Group 1 was the ischemic control group. Group 2 was treated with enoant of 1.25 g kg⁻¹ per day for 15 days after I/R. Group 3 received the same concentration of enoant as in group 2 for 15 days before and after I/R. Group 4 was the sham operation group. EEG activities were recorded and the levels of TNF-α, IL-1ß and IL-6, TBARS and GSH were measured in the whole brain homogenate. RESULTS: There were significant changes in EEG activity in groups treated with enoant either before or after ischemia when compared with their basal EEG values. TNF-α, IL-6 and IL-1ß levels were significantly increased after I/R. GSH levels in group 3 treated with enoant in both pre- and post-ischemic periods were significantly increased and TBARS concentration was decreased compared with the ischemic group. CONCLUSION: The findings support that both pre-ischemic and post-ischemic administrations of enoant might produce neuroprotective action against cerebral ischemia.


Assuntos
Antioxidantes/farmacologia , Bebidas , Isquemia Encefálica/tratamento farmacológico , Eletroencefalografia/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Vitis/química , Animais , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/prevenção & controle , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Reperfusão , Traumatismo por Reperfusão/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA