Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Neurocrit Care ; 39(1): 218-228, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37349601

RESUMO

BACKGROUND: Aneurismal subarachnoid hemorrhage (SAH) is a type of hemorrhagic stroke that, despite improvement through therapeutic interventions, remains a devastating cerebrovascular disorder that has a high mortality rate and causes long-term disability. Cerebral inflammation after SAH is promoted through microglial accumulation and phagocytosis. Furthermore, proinflammatory cytokine release and neuronal cell death play key roles in the development of brain injury. The termination of these inflammation processes and restoration of tissue homeostasis are of utmost importance regarding the possible chronicity of cerebral inflammation and the improvement of the clinical outcome for affected patients post SAH. Thus, we evaluated the inflammatory resolution phase post SAH and considered indications for potential tertiary brain damage in cases of incomplete resolution. METHODS: Subarachnoid hemorrhage was induced through endovascular filament perforation in mice. Animals were killed 1, 7 and 14 days and 1, 2 and 3 months after SAH. Brain cryosections were immunolabeled for ionized calcium-binding adaptor molecule-1 to detect microglia/macrophages. Neuronal nuclei and terminal deoxyuridine triphosphate-nick end labeling staining was used to visualize secondary cell death of neurons. The gene expression of various proinflammatory mediators in brain samples was analyzed by quantitative polymerase chain reaction. RESULTS: We observed restored tissue homeostasis due to decreased microglial/macrophage accumulation and neuronal cell death 1 month after insult. However, the messenger RNA expression levels of  interleukin 6  and  tumor necrosis factor α were still elevated at 1 and 2 months post SAH, respectively. The gene expression of interleukin 1ß reached its maximum on day 1, whereas at later time points, no significant differences between the groups were detected. CONCLUSIONS: By the herein presented molecular and histological data we provide an important indication for an incomplete resolution of inflammation within the brain parenchyma after SAH. Inflammatory resolution and the return to tissue homeostasis represent an important contribution to the disease's pathology influencing the impact on brain damage and outcome after SAH. Therefore, we consider a novel complementary or even superior therapeutic approach that should be carefully rethought in the management of cerebral inflammation after SAH. An acceleration of the resolution phase at the cellular and molecular levels could be a potential aim in this context.


Assuntos
Lesões Encefálicas , Hemorragia Subaracnóidea , Camundongos , Animais , Hemorragia Subaracnóidea/tratamento farmacológico , Inflamação/etiologia , Inflamação/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças
2.
Neurocrit Care ; 37(2): 410-423, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35585424

RESUMO

BACKGROUND: Neuroprotective treatment strategies aiming at interfering with either inflammation or cell death indicate the importance of these mechanisms in the development of brain injury after subarachnoid hemorrhage (SAH). This study was undertaken to evaluate the influence of minocycline on microglia/macrophage cell activity and its neuroprotective and anti-inflammatory impact 14 days after aneurismal SAH in mice. METHODS: Endovascular filament perforation was used to induce SAH in mice. SAH + vehicle-operated mice were used as controls for SAH vehicle-treated mice and SAH + minocycline-treated mice. The drug administration started 4 h after SAH induction and was daily repeated until day 7 post SAH and continued until day 14 every second day. Brain cryosections were immunolabeled for Iba1 to detect microglia/macrophages and NeuN to visualize neurons. Phagocytosis assay was performed to determine the microglia/macrophage activity status. Apoptotic cells were stained using terminal deoxyuridine triphosphate nick end labeling. Real-time quantitative polymerase chain reaction was used to estimate cytokine gene expression. RESULTS: We observed a significantly reduced phagocytic activity of microglia/macrophages accompanied by a lowered spatial interaction with neurons and reduced neuronal apoptosis achieved by minocycline administration after SAH. Moreover, the SAH-induced overexpression of pro-inflammatory cytokines and neuronal cell death was markedly attenuated by the compound. CONCLUSIONS: Minocycline treatment may be implicated as a therapeutic approach with long-term benefits in the management of secondary brain injury after SAH in a clinically relevant time window.


Assuntos
Lesões Encefálicas , Fármacos Neuroprotetores , Hemorragia Subaracnóidea , Animais , Anti-Inflamatórios/farmacologia , Apoptose , Lesões Encefálicas/complicações , Morte Celular , Citocinas/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos , Camundongos , Microglia/metabolismo , Minociclina/farmacologia , Minociclina/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/complicações
3.
J Neuroinflammation ; 18(1): 36, 2021 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-33516246

RESUMO

BACKGROUND: Microglia-driven cerebral spreading inflammation is a key contributor to secondary brain injury after SAH. Genetic depletion or deactivation of microglia has been shown to ameliorate neuronal cell death. Therefore, clinically feasible anti-inflammatory approaches counteracting microglia accumulation or activation are interesting targets for SAH treatment. Here, we tested two different methods of interference with microglia-driven cerebral inflammation in a murine SAH model: (i) inflammatory preconditioning and (ii) pharmacological deactivation. METHODS: 7T-MRI-controlled SAH was induced by endovascular perforation in four groups of C57Bl/6 mice: (i) Sham-operation, (ii) SAH naïve, (iii) SAH followed by inflammatory preconditioning (LPS intraperitoneally), and (iv) SAH followed by pharmacological microglia deactivation (colony-stimulating factor-1 receptor-antagonist PLX3397 intraperitoneally). Microglia accumulation and neuronal cell death (immuno-fluorescence), as well as activation status (RT-PCR for inflammation-associated molecules from isolated microglia) were recorded at day 4 and 14. Toll-like receptor4 (TLR4) status was analyzed using FACS. RESULTS: Following SAH, significant cerebral spreading inflammation occurred. Microglia accumulation and pro-inflammatory gene expression were accompanied by neuronal cell death with a maximum on day 14 after SAH. Inflammatory preconditioning as well as PLX3397-treatment resulted in significantly reduced microglia accumulation and activation as well as neuronal cell death. TLR4 surface expression in preconditioned animals was diminished as a sign for receptor activation and internalization. CONCLUSIONS: Microglia-driven cerebral spreading inflammation following SAH contributes to secondary brain injury. Two microglia-focused treatment strategies, (i) inflammatory preconditioning with LPS and (ii) pharmacological deactivation with PLX3397, led to significant reduction of neuronal cell death. Increased internalization of inflammation-driving TLR4 after preconditioning leaves less receptor molecules on the cell surface, providing a probable explanation for significantly reduced microglia activation. Our findings support microglia-focused treatment strategies to overcome secondary brain injury after SAH. Delayed inflammation onset provides a valuable clinical window of opportunity.


Assuntos
Anti-Inflamatórios/administração & dosagem , Lesões Encefálicas/metabolismo , Lesões Encefálicas/prevenção & controle , Microglia/metabolismo , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/metabolismo , Aminopiridinas/administração & dosagem , Animais , Lesões Encefálicas/diagnóstico por imagem , Precondicionamento Isquêmico/métodos , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Pirróis/administração & dosagem , Hemorragia Subaracnóidea/diagnóstico por imagem
4.
Int J Mol Sci ; 22(20)2021 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-34681839

RESUMO

Tumor recurrence is the main challenge in glioblastoma (GBM) treatment. Gold standard therapy temozolomide (TMZ) is known to induce upregulation of IL8/CXCL2/CXCR2 signaling that promotes tumor progression and angiogenesis. Our aim was to verify the alterations on this signaling pathway in human GBM recurrence and to investigate the impact of TMZ in particular. Furthermore, a combi-therapy of TMZ and CXCR2 antagonization was established to assess the efficacy and tolerability. First, we analyzed 76 matched primary and recurrent GBM samples with regard to various histological aspects with a focus on the role of TMZ treatment and the assessment of predictors of overall survival (OS). Second, the combi-therapy with TMZ and CXCR2-antagonization was evaluated in a syngeneic mouse tumor model with in-depth immunohistological investigations and subsequent gene expression analyses. We observed a significantly decreased infiltration of tumor-associated microglia/macrophages (TAM) in recurrent tumors, while a high TAM infiltration in primary tumors was associated with a reduced OS. Additionally, more patients expressed IL8 in recurrent tumors and TMZ therapy maintained CXCL2 expression. In mice, enhanced anti-tumoral effects were observed after combi-therapy. In conclusion, high TAM infiltration predicts a survival disadvantage, supporting findings of the tumor-promoting phenotype of TAMs. Furthermore, the combination therapy seemed to be promising to overcome CXCR2-mediated resistance.


Assuntos
Glioblastoma/metabolismo , Recidiva Local de Neoplasia/metabolismo , Compostos de Fenilureia/farmacologia , Receptores de Interleucina-8B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Temozolomida/farmacologia , Macrófagos Associados a Tumor/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos Alquilantes/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Encefálicas/metabolismo , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-8/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Neovascularização Patológica/fisiopatologia , Prognóstico , Análise de Sobrevida , Microambiente Tumoral/efeitos dos fármacos , Adulto Jovem
5.
Cell Mol Neurobiol ; 40(5): 765-766, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32424770

RESUMO

The original version of the article unfortunately contained an error in the unit of the protein concentrations under 'Stereotactic Intraparenchymal Injections' subsection in 'Methods' section.

6.
Cell Mol Neurobiol ; 40(5): 751-764, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31858356

RESUMO

Pigment epithelium-derived factor (PEDF) is a neurotrophic factor with neuroprotective, antiangiogenic, and antipermeability effects. In the brain, blood-brain barrier (BBB) function is essential for homeostasis. Its impairment plays a crucial role in the pathophysiology of many neurological diseases, including ischemic stroke. We investigated (a) whether PEDF counteracted vascular endothelial growth factor (VEGF)-induced BBB disruption in the mouse brain, (b) the time course and route of BBB permeability and the dynamics of PEDF expression after cerebral ischemia, and (c) whether intraventricular infusion of PEDF ameliorated brain ischemia by reducing BBB impairment. C57Bl6/N mice received intraparenchymal injections of CSF, VEGF, or a combination of VEGF and PEDF. PEDF increased paracellular but not transcellular BBB integrity as indicated by an increase in the tight junction protein claudin-5. In another group of mice undergoing 60-min middle cerebral artery occlusion (MCAO), transcellular BBB permeability (fibrinogen staining in the absence of a loss of claudin-5) increased as early as 6 h after reperfusion. PEDF immunofluorescence increased at 24 h, which paralleled with a decreased paracellular BBB permeability (claudin-5). PEDF after MCAO originated from the blood stream and endogenous pericytes. In the third experiment, the intraventricular infusion of PEDF decreased edema and cell death after MCAO, potentially mediated by the improvement of the paracellular route of BBB permeability (claudin-5) in the absence of an amelioration of Evans Blue extravasation. Together, our data suggest that PEDF improves BBB function after cerebral ischemia by affecting the paracellular but not the transcellular route. However, further quantitative data of the different routes of BBB permeability will be required to validate our findings.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Proteínas do Olho/farmacologia , Ataque Isquêmico Transitório/terapia , Fatores de Crescimento Neural/farmacologia , Fármacos Neuroprotetores/farmacologia , Serpinas/farmacologia , Animais , Barreira Hematoencefálica/lesões , Barreira Hematoencefálica/metabolismo , Claudina-5/metabolismo , Modelos Animais de Doenças , Proteínas do Olho/uso terapêutico , Ataque Isquêmico Transitório/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Serpinas/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/farmacologia
7.
Arterioscler Thromb Vasc Biol ; 37(5): 867-878, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28254815

RESUMO

OBJECTIVE: Cerebral edema caused by the disruption of the blood-brain barrier is a major complication after stroke. Therefore, strategies to accelerate and enhance neurovascular recovery after stroke are of prime interest. Our main aim was to study the role of ephrinB2/EphB4 signaling in mediating the vascular repair and in blood-brain barrier restoration after mild cerebral ischemia occlusion/reperfusion. APPROACH AND RESULTS: Here, we show that the guidance molecule ephrinB2 plays a key role in neurovascular protection and blood-brain barrier restoration after stroke. In a focal stroke model, we characterize the stroke-induced damage to cerebral blood vessels and their subsequent endogenous repair on a cellular, molecular, and functional level. EphrinB2 and its tyrosine kinase receptor EphB4 are upregulated early after stroke by endothelial cells and perivascular support cells, in parallel to their reassembly during neurovascular recovery. Using both retroviral and pharmacological approaches, we show that the inhibition of ephrinB2/EphB4 signaling suppresses post-middle cerebral artery occlusion neurovascular repair mechanisms resulting in an aggravation of brain swelling. In contrast, the activation of ephrinB2 after brain ischemia leads to an increased pericyte recruitment and increased endothelial-pericyte interaction, resulting in an accelerated neurovascular repair after ischemia. CONCLUSIONS: We show that reducing swelling could result in improved outcome because of reduction in damaged brain tissue. We also identify a novel role for ephrinB2/EphB4 signaling in the maintenance of the neurovascular homeostasis and provide a novel therapeutic approach in reducing brain swelling after stroke.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Edema Encefálico/prevenção & controle , Efrina-B2/agonistas , Terapia Genética , Infarto da Artéria Cerebral Média/terapia , Neovascularização Fisiológica/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Edema Encefálico/genética , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Linhagem Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Efrina-B2/genética , Efrina-B2/metabolismo , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Pericitos/metabolismo , Pericitos/patologia , Fosforilação , Interferência de RNA , Receptor EphB4/genética , Receptor EphB4/metabolismo , Índice de Gravidade de Doença , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
8.
Circ Res ; 115(6): 581-90, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25057127

RESUMO

RATIONALE: Endothelial cell-specific molecule 1 (Esm1) is a secreted protein thought to play a role in angiogenesis and inflammation. However, there is currently no direct in vivo evidence supporting a function of Esm1 in either of these processes. OBJECTIVE: To determine the role of Esm1 in vivo and the underlying molecular mechanisms. METHODS AND RESULTS: We generated and analyzed Esm1 knockout (Esm1(KO)) mice to study its role in angiogenesis and inflammation. Esm1 expression is induced by the vascular endothelial growth factor A (VEGF-A) in endothelial tip cells of the mouse retina. Esm1(KO) mice showed delayed vascular outgrowth and reduced filopodia extension, which are both VEGF-A-dependent processes. Impairment of Esm1 function led to a decrease in phosphorylated Erk1/2 (extracellular-signal regulated kinases 1/2) in sprouting vessels. We also found that Esm1(KO) mice displayed a 40% decrease in leukocyte transmigration. Moreover, VEGF-induced vascular permeability was decreased by 30% in Esm1(KO) mice and specifically on stimulation with VEGF-A165 but not VEGF-A121. Accordingly, cerebral edema attributable to ischemic stroke-induced vascular permeability was reduced by 50% in the absence of Esm1. Mechanistically, we show that Esm1 binds directly to fibronectin and thereby displaces fibronectin-bound VEGF-A165 leading to increased bioavailability of VEGF-A165 and subsequently enhanced levels of VEGF-A signaling. CONCLUSIONS: Esm1 is simultaneously a target and modulator of VEGF signaling in endothelial cells, playing a role in angiogenesis, inflammation, and vascular permeability, which might be of potential interest for therapeutic applications.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Membrana Celular/fisiologia , Células Endoteliais/fisiologia , Proteoglicanas/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Disponibilidade Biológica , Fibronectinas/metabolismo , Inflamação/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Neovascularização Fisiológica/fisiologia , Proteoglicanas/deficiência , Proteoglicanas/genética , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Brain Spine ; 2: 100859, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36248104

RESUMO

Introduction: Traumatic Spinal Cord Injury (SCI) is one of the leading causes of disability in the world. Treatment is limited to supportive care and no curative therapy exists. Experimental research to understand the complex pathophysiology and potential mediators of spinal cord regeneration is essential to develop innovative translational therapies. A multitude of experimental imaging methods to monitor spinal cord regeneration in vivo have developed over the last years. However, little literature exists to deal with advanced imaging methods specifically available in SCI research. Research Question: This systematic literature review examines the current standards in experimental imaging in SCI allowing for in vivo imaging of spinal cord regeneration on a neuronal, vascular, and cellular basis. Material and Methods: Articles were included meeting the following criteria: experimental research, original studies, rodent subjects, and intravital imaging. Reviewed in detail are microstructural and functional Magnetic Resonance Imaging, Micro-Computed Tomography, Laser Speckle Imaging, Very High Resolution Ultrasound, and in vivo microscopy techniques. Results: Following the PRISMA guidelines for systematic reviews, 689 articles were identified for review, of which 492 were sorted out after screening and an additional 104 after detailed review. For qualitative synthesis 93 articles were included in this publication. Discussion and Conclusion: With this study we give an up-to-date overview about modern experimental imaging techniques with the potential to advance the knowledge on spinal cord regeneration following SCI. A thorough knowledge of the strengths and limitations of the reviewed techniques will help to optimally exploit our current experimental armamentarium in the field.

10.
PLoS One ; 17(4): e0266319, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35446876

RESUMO

Interstitial fibrosis and tubular atrophy, a major cause of kidney allograft dysfunction, has been linked to premature cellular senescence. The mTOR inhibitor Rapamycin protects from senescence in experimental models, but its antiproliferative properties have raised concern early after transplantation particularly at higher doses. Its effect on senescence has not been studied in kidney transplantation, yet. Rapamycin was applied to a rat kidney transplantation model (3 mg/kg bodyweight loading dose, 1.5 mg/kg bodyweight daily dose) for 7 days. Low Rapamycin trough levels (2.1-6.8 ng/mL) prevented the accumulation of p16INK4a positive cells in tubules, interstitium, and glomerula. Expression of the cytokines MCP-1, IL-1ß, and TNF-α, defining the proinflammatory senescence-associated secretory phenotype, was abrogated. Infiltration with monocytes/macrophages and CD8+ T-lymphocytes was reduced and tubular function was preserved by Rapamycin. Inhibition of mTOR was not associated with impaired structural recovery, higher glucose levels, or weight loss. mTOR inhibition with low-dose Rapamycin in the immediate posttransplant period protected from premature cellular senescence without negative effects on structural and functional recovery from preservation/reperfusion damage, glucose homeostasis, and growth in a rat kidney transplantation model. Reduced senescence might maintain the renal regenerative capacity rendering resilience to future injuries resulting in protection from interstitial fibrosis and tubular atrophy.


Assuntos
Nefropatias , Transplante de Rim , Animais , Atrofia/patologia , Senescência Celular , Feminino , Fibrose , Glucose/farmacologia , Humanos , Rim/patologia , Nefropatias/patologia , Transplante de Rim/efeitos adversos , Masculino , Ratos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
11.
J Cereb Blood Flow Metab ; 42(1): 121-135, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34427143

RESUMO

Physiological effects of spreading depolarizations (SD) are only well studied in the first hours after experimental stroke. In patients with malignant hemispheric stroke (MHS), monitoring of SDs is restricted to the postoperative ICU stay, typically day 2-7 post-ictus. Therefore, we investigated the role of physiological variables (temperature, intracranial pressure, mean arterial pressure and cerebral perfusion pressure) in relationship to SD during the late phase after MHS in humans. Additionally, an experimental stroke model was used to investigate hemodynamic consequences of SD during this time window. In 60 patients with MHS, the occurrence of 1692 SDs was preceded by a decrease in mean arterial pressure (-1.04 mmHg; p = .02) and cerebral perfusion pressure (-1.04 mmHg; p = .03). Twenty-four hours after middle cerebral artery occlusion in 50 C57Bl6/J mice, hypothermia led to prolonged SD-induced hyperperfusion (+2.8 min; p < .05) whereas hypertension mitigated initial hypoperfusion (-1.4 min and +18.5%Δ rCBF; p < .01). MRI revealed that SDs elicited 24 hours after experimental stroke were associated with lesion progression (15.9 vs. 14.8 mm³; p < .01). These findings of small but significant effects of physiological variables on SDs in the late phase after ischemia support the hypothesis that the impact of SDs may be modified by adjusting physiological variables.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical , AVC Isquêmico/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Estudos Prospectivos
12.
Sci Rep ; 11(1): 16270, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381142

RESUMO

mTOR inhibitors offer advantages after kidney transplantation including antiviral and antitumor activity besides facilitating low calcineurin inhibitor exposure to reduce nephrotoxicity. Concerns about adverse effects due to antiproliferative and antiangiogenic properties have limited their clinical use particularly early after transplantation. Interference with vascular endothelial growth factor (VEGF)-A, important for physiologic functioning of renal endothelial cells and tubular epithelium, has been implicated in detrimental renal effects of mTOR inhibitors. Low doses of Rapamycin (loading dose 3 mg/kg bodyweight, daily doses 1.5 mg/kg bodyweight) were administered in an allogenic rat kidney transplantation model resulting in a mean through concentration of 4.30 ng/mL. Glomerular and peritubular capillaries, tubular cell proliferation, or functional recovery from preservation/reperfusion injury were not compromised in comparison to vehicle treated animals. VEGF-A, VEGF receptor 2, and the co-receptor Neuropilin-1 were upregulated by Rapamycin within 7 days. Rat proximal tubular cells (RPTC) responded in vitro to hypoxia with increased VEGF-A and VEGF-R1 expression that was not suppressed by Rapamycin at therapeutic concentrations. Rapamycin did not impair proliferation of RPTC under hypoxic conditions. Low-dose Rapamycin early posttransplant does not negatively influence the VEGF network crucial for recovery from preservation/reperfusion injury. Enhancement of VEGF signaling peritransplant holds potential to further improve outcomes.


Assuntos
Transplante de Rim , Túbulos Renais Proximais/irrigação sanguínea , Túbulos Renais Proximais/fisiologia , Resultados Negativos , Regeneração/efeitos dos fármacos , Sirolimo/efeitos adversos , Animais , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Túbulos Renais Proximais/citologia , Masculino , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Traumatismo por Reperfusão/genética , Sirolimo/administração & dosagem , Sirolimo/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
13.
J Vis Exp ; (178)2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34978299

RESUMO

The endovascular filament perforation model to mimic subarachnoid hemorrhage (SAH) is a commonly used model - however, the technique can cause a high mortality rate as well as an uncontrollable volume of SAH and other intracranial complications such as stroke or intracranial hemorrhage. In this protocol, a standardized SAH mouse model is presented, induced by endovascular filament perforation, combined with magnetic resonance imaging (MRI) 24 h after operation to ensure the correct bleeding site and exclude other relevant intracranial pathologies. Briefly, C57BL/6J mice are anesthetized with an intraperitoneal ketamine/xylazine (70 mg/16 mg/kg body weight) injection and placed in a supine position. After midline neck incision, the common carotid artery (CCA) and carotid bifurcation are exposed, and a 5-0 non-absorbable monofilament polypropylene suture is inserted in a retrograde fashion into the external carotid artery (ECA) and advanced into the common carotid artery. Then, the filament is invaginated into the internal carotid artery (ICA) and pushed forward to perforate the anterior cerebral artery (ACA). After recovery from surgery, mice undergo a 7.0 T MRI 24 h later. The volume of bleeding can be quantified and graded via postoperative MRI, enabling a robust experimental SAH group with the option to perform further subgroup analyses based on blood quantity.


Assuntos
Hemorragia Subaracnóidea , Animais , Artéria Carótida Interna , Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Hemorragia Subaracnóidea/diagnóstico por imagem , Hemorragia Subaracnóidea/etiologia , Hemorragia Subaracnóidea/cirurgia
14.
Cancers (Basel) ; 13(14)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34298631

RESUMO

CyberKnife stereotactic radiosurgery (CK-SRS) precisely delivers radiation to intracranial tumors. However, the underlying radiobiological mechanisms at high single doses are not yet fully understood. Here, we established and evaluated the early radiobiological effects of CK-SRS treatment at a single dose of 20 Gy after 15 days of tumor growth in a syngeneic glioblastoma-mouse model. Exact positioning was ensured using a custom-made, non-invasive, and trackable frame. One superimposed target volume for the CK-SRS planning was created from the fused tumor volumes obtained from MRIs prior to irradiation. Dose calculation and delivery were planned using a single-reference CT scan. Six days after irradiation, tumor volumes were measured using MRI scans, and radiobiological effects were assessed using immunofluorescence staining. We found that CK-SRS treatment reduced tumor volume by approximately 75%, impaired cell proliferation, diminished tumor vasculature, and increased immune response. The accuracy of the delivered dose was demonstrated by staining of DNA double-strand breaks in accordance with the planned dose distribution. Overall, we confirmed that our proposed setup enables the precise irradiation of intracranial tumors in mice using only one reference CT and superimposed MRI volumes. Thus, our proposed mouse model for reproducible CK-SRS can be used to investigate radiobiological effects and develop novel therapeutic approaches.

15.
Front Neurosci ; 15: 756577, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899163

RESUMO

Purpose: Subsurface blood vessels in the cerebral cortex have been identified as a bottleneck in cerebral perfusion with the potential for collateral remodeling. However, valid techniques for non-invasive, longitudinal characterization of neocortical microvessels are still lacking. In this study, we validated contrast-enhanced magnetic resonance imaging (CE-MRI) for in vivo characterization of vascular changes in a model of spontaneous collateral outgrowth following chronic cerebral hypoperfusion. Methods: C57BL/6J mice were randomly assigned to unilateral internal carotid artery occlusion or sham surgery and after 21 days, CE-MRI based on T2*-weighted imaging was performed using ultra-small superparamagnetic iron oxide nanoparticles to obtain subtraction angiographies and steady-state cerebral blood volume (ss-CBV) maps. First pass dynamic susceptibility contrast MRI (DSC-MRI) was performed for internal validation of ss-CBV. Further validation at the histological level was provided by ex vivo serial two-photon tomography (STP). Results: Qualitatively, an increase in vessel density was observed on CE-MRI subtraction angiographies following occlusion; however, a quantitative vessel tracing analysis was prone to errors in our model. Measurements of ss-CBV reliably identified an increase in cortical vasculature, validated by DSC-MRI and STP. Conclusion: Iron oxide nanoparticle-based ss-CBV serves as a robust, non-invasive imaging surrogate marker for neocortical vessels, with the potential to reduce and refine preclinical models targeting the development and outgrowth of cerebral collateralization.

16.
Eur J Cancer ; 126: 106-115, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31927212

RESUMO

OBJECTIVE: Besides VEGF, alternative signalling via CXCR2 and its ligands CXCL2/CXCL8 is a crucial part of angiogenesis in glioblastoma. Our aim was to understand the role of CXCR2 for glioma biology and elucidate the therapeutic potential of its specific inhibition. METHODS: GL261 glioma cells were implanted intracranially in syngeneic mice. The 14 or 7 days of local or systemic treatment with CXCR2-antagonist (SB225002) was initiated early on the day of tumour cell implantation or delayed after 14 days of tumour growth. Glioma volume was verified using MRI before and after treatment. Immunofluorescence staining was used to investigate tumour progression, angiogenesis and microglial behaviour. Furthermore, in vitro assays and gene expression analyses of glioma and endothelial cells were performed to validate inhibitor activity. RESULTS: CXCR2-blocking led to significantly reduced glioma volumes of around 50% after early and delayed local treatments. The treated tumours were comparable with controls regarding invasiveness, proliferation and apoptotic cell activity. Furthermore, no differences in CXCR2/CXCL2 expression were observed. However, immunostaining revealed reduction in vessel density and accumulation of microglia/macrophages, whereas interaction of these myeloid cells with tumour vessels was enhanced. In vitro analyses of the CXCR2-antagonist showed its direct impact on proliferation of glioma and endothelial cells if used at higher concentrations. In addition, expression of CXCR2/CXCL2 signalling genes was increased in both cell types by SB225002, but VEGF-relevant genes were unaffected. CONCLUSION: The CXCR2-antagonist inhibited glioma growth during tumour initiation and progression, whereas treatment was well-tolerated by the recipients. Thus, the CXCR2/CXCL2 signalling represents a promising therapeutic target in glioma.


Assuntos
Neoplasias Encefálicas/prevenção & controle , Quimiocina CXCL2/metabolismo , Glioma/prevenção & controle , Compostos de Fenilureia/farmacologia , Receptores de Interleucina-8B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Glioma/irrigação sanguínea , Glioma/metabolismo , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Imageamento por Ressonância Magnética , Camundongos Endogâmicos C57BL , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/metabolismo , Neovascularização Patológica/prevenção & controle , Receptores de Interleucina-8B/metabolismo , Carga Tumoral/efeitos dos fármacos
17.
Acta Biomater ; 101: 395-402, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31726251

RESUMO

Respiratory arrest is a major life-threatening condition leading to cessation of vital functions and hypoxic-anoxic injury of the brain. The progressive structural tissue changes characterizing the dying brain biophysically are unknown. Here we use noninvasive magnetic resonance elastography to show that biomechanical tissue properties are highly sensitive to alterations in the brain in the critical period before death. Our findings demonstrate that brain stiffness increases after respiratory arrest even when cardiac function is still preserved. Within 5 min of cardiac arrest, cerebral stiffness further increases by up to 30%. This early mechanical signature of the dying brain can be explained by water accumulation and redistribution from extracellular spaces into cells. These processes, together, increase interstitial and intracellular pressure as revealed by magnetic resonance spectroscopy and diffusion-weighted imaging. Our data suggest that the fast response of cerebral stiffness to respiratory arrest enables the monitoring of life-threatening brain pathology using noninvasive in vivo imaging. STATEMENT OF SIGNIFICANCE: Hypoxia-anoxia is a life-threatening condition eventually leading to brain death. Therefore, monitoring vital brain functions in patients at risk is urgently required during emergency care or treatment of acute brain damage due to insufficient oxygen supply. In mouse model of hypoxia-anoxia, we have shown for the first time that biophysical tissue parameters such as brain stiffness changed markedly during the process of death.


Assuntos
Morte Encefálica/diagnóstico por imagem , Encéfalo/fisiopatologia , Técnicas de Imagem por Elasticidade , Hipóxia/fisiopatologia , Imageamento por Ressonância Magnética , Animais , Fenômenos Biomecânicos , Camundongos Endogâmicos C57BL
18.
Kidney Int ; 75(7): 699-710, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19190681

RESUMO

Glomerulonephritis is characterized by hematuria, proteinuria, hypertension, and edema, but the mechanisms contributing to volume disorders are controversial. Here we used the rat anti-Thy1 model of mesangioproliferative glomerulonephritis to test the hypothesis that disturbed salt and water homeostasis is based on tubular epithelial changes that cause salt retention. In this model there was an early onset of pronounced proteinuria and lipiduria associated with reduced fractional sodium excretion and a lowering of the renin-angiotensin-aldosterone system. The glomerular filtration rate and creatinine clearance were decreased on day 6. There was a reduced abundance of the major salt and water transport proteins on the proximal tubular brush border membrane and which paralleled cellular protein overload, enhanced membrane cholesterol uptake and cytoskeletal changes. Alterations in thick ascending limb were moderate. Changes in the collecting ducts were characterized by an enhanced abundance and increased subunit cleavage of the epithelial sodium channel, both events consistent with increased sodium reabsorption. We suggest that irrespective of the proximal tubular changes, altered collecting duct sodium reabsorption may be crucial for volume retention in acute glomerulonephritis. We suggest that enhanced proteolytic cleavage of ion transporter subunits might be a novel mechanism of channel activation in glomerular diseases. Whether these proteases are filtered or locally secreted awaits determination.


Assuntos
Glomerulonefrite/fisiopatologia , Túbulos Renais Proximais/metabolismo , Equilíbrio Hidroeletrolítico , Animais , Proteínas de Transporte/análise , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Canais Epiteliais de Sódio/análise , Canais Epiteliais de Sódio/metabolismo , Glomerulonefrite/imunologia , Túbulos Renais Proximais/patologia , Ratos , Sódio/metabolismo
19.
J Neurosurg ; 132(2): 465-472, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30684943

RESUMO

OBJECTIVE: The management of patients with aneurysmal subarachnoid hemorrhage (aSAH) remains a highly demanding challenge in critical care medicine. Despite all efforts, the calcium channel antagonist nimodipine remains the only drug approved for improving outcomes after aSAH. However, in its current form of application, it provides less than optimal efficacy and causes dose-limiting hypotension in a substantial number of patients. Here, the authors tested in vitro the release dynamics of a novel formulation of the calcium channel blocker nicardipine and in vivo local tolerance and tissue reaction using a chronic cranial window model in mice. METHODS: To characterize the release kinetics in vitro, dissolution experiments were performed using artificial cerebrospinal fluid over a time period of 21 days. The excipients used in this formulation (NicaPlant) for sustained nicardipine release are a mixture of two completely degradable polymers. A chronic cranial window in C57BL/6 mice was prepared, and NicaPlant slices were placed in proximity to the exposed cerebral vasculature. Epifluorescence video microscopy was performed right after implantation and on days 3 and 7 after surgery. Vessel diameter of the arteries and veins, vessel permeability, vessel configuration, and leukocyte-endothelial cell interaction were quantified by computer-assisted analysis. Immunofluorescence staining was performed to analyze inflammatory reactions and neuronal alterations. RESULTS: In vitro the nicardipine release profile showed an almost linear curve with about 80% release at day 15 and full release at day 21. In vivo epifluorescence video microscopy showed a significantly higher arterial vessel diameter in the NicaPlant group due to vessel dilatation (21.6 ± 2.6 µm vs 17.8 ± 1.5 µm in controls, p < 0.01) confirming vasoactivity of the implant, whereas the venous diameter was not affected. Vessel dilatation did not have any influence on the vessel permeability measured by contrast extravasation of the fluorescent dye in epifluorescence microscopy. Further, an increased leukocyte-endothelial cell interaction due to the implant could not be detected. Histological analysis did not show any microglial activation or accumulation. No structural neuronal changes were observed. CONCLUSIONS: NicaPlant provides continuous in vitro release of nicardipine over a 3-week observation period. In vivo testing confirmed vasoactivity and lack of toxicity. The local application of this novel nicardipine delivery system to the subarachnoid space is a promising tool to improve patient outcomes while avoiding systemic side effects.


Assuntos
Encéfalo/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Nicardipino/administração & dosagem , Hemorragia Subaracnóidea/tratamento farmacológico , Animais , Encéfalo/metabolismo , Bloqueadores dos Canais de Cálcio/metabolismo , Preparações de Ação Retardada , Avaliação Pré-Clínica de Medicamentos/métodos , Implantes de Medicamento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nicardipino/metabolismo , Hemorragia Subaracnóidea/metabolismo
20.
Nat Commun ; 10(1): 2817, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31249304

RESUMO

Sufficient vascular supply is indispensable for brain development and function, whereas dysfunctional blood vessels are associated with human diseases such as vascular malformations, stroke or neurodegeneration. Pericytes are capillary-associated mesenchymal cells that limit vascular permeability and protect the brain by preserving blood-brain barrier integrity. Loss of pericytes has been linked to neurodegenerative changes in genetically modified mice. Here, we report that postnatal inactivation of the Rbpj gene, encoding the transcription factor RBPJ, leads to alteration of cell identity markers in brain pericytes, increases local TGFß signalling, and triggers profound changes in endothelial behaviour. These changes, which are not mimicked by pericyte ablation, imperil vascular stability and induce the acquisition of pathological landmarks associated with cerebral cavernous malformations. In adult mice, loss of Rbpj results in bigger stroke lesions upon ischemic insult. We propose that brain pericytes can acquire deleterious properties that actively enhance vascular lesion formation and promote pathogenic processes.


Assuntos
Encéfalo/metabolismo , Hemangioma Cavernoso do Sistema Nervoso Central/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Pericitos/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/citologia , Progressão da Doença , Feminino , Hemangioma Cavernoso do Sistema Nervoso Central/genética , Hemangioma Cavernoso do Sistema Nervoso Central/patologia , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Masculino , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA