Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sensors (Basel) ; 24(13)2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-39000864

RESUMO

Quartz-Enhanced Photoacoustic Spectroscopy (QEPAS) is a technique in which the sound wave is detected by a quartz tuning fork (QTF). It enables particularly high specificity with respect to the excitation frequency and is well known for an extraordinarily sensitive analysis of gaseous samples. We have developed the first photoacoustic (PA) cell for QEPAS on solid samples. Periodic heating of the sample is excited by modulated light from an interband cascade laser (ICL) in the infrared region. The cell represents a half-open cylinder that exhibits an acoustical resonance frequency equal to that of the QTF and, therefore, additionally amplifies the PA signal. The antinode of the sound pressure of the first longitudinal overtone can be accessed by the sound detector. A 3D finite element (FE) simulation confirms the optimal dimensions of the new cylindrical cell with the given QTF resonance frequency. An experimental verification is performed with an ultrasound micro-electromechanical system (MEMS) microphone. The presented frequency-dependent QEPAS measurement exhibits a low noise signal with a high-quality factor. The QEPAS-based investigation of three different solid synthetics resulted in a linearly dependent signal with respect to the absorption.

2.
Sci Rep ; 14(1): 2441, 2024 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-38286816

RESUMO

Traumatic brain injury (TBI) is a leading cause of mortality and disability worldwide. Acute neuroinflammation is a prominent reaction after TBI and is mostly initiated by brain-resident glial cells such as microglia, NG2-glia and astrocytes. The magnitude of this reaction paves the way for long-lasting consequences such as chronic neurological pathologies, for which therapeutic options remain limited. The neuroinflammatory response to TBI is mostly studied with craniotomy-based animal models that are very robust but also rather artificial. Here, we aimed to analyze the reaction of glial cells in a highly translational but variable closed head injury (CHI) model and were able to correlate the severity of the trauma to the degree of glial response. Furthermore, we could show that the different glial cell types react in a temporally and spatially orchestrated manner in terms of morphological changes, proliferation, and cell numbers in the first 15 days after the lesion. Interestingly, NG2-glia, the only proliferating cells in the healthy brain parenchyma, divided at a rate that was correlated with the size of the injury. Our findings describe the previously uncharacterized posttraumatic response of the major brain glial cell types in CHI in order to gain a detailed understanding of the course of neuroinflammatory events; such knowledge may open novel avenues for future therapeutic approaches in TBI.


Assuntos
Lesões Encefálicas Traumáticas , Traumatismos Cranianos Fechados , Animais , Neuroglia/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas Traumáticas/patologia , Astrócitos/metabolismo , Microglia/metabolismo , Traumatismos Cranianos Fechados/patologia , Modelos Animais de Doenças
3.
Aging Cell ; 22(9): e13911, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37609868

RESUMO

Aging of the central nervous system (CNS) leads to motoric and cognitive decline and increases the probability for neurodegenerative disease development. Astrocytes fulfill central homeostatic functions in the CNS including regulation of immune responses and metabolic support of neurons and oligodendrocytes. In this study, we investigated the effect of redox imbalance in astrocytes by using a conditional astrocyte-specific SOD2-deficient mouse model (SOD2ako ) and analyzed these animals at different stages of their life. SOD2ako mice did not exhibit any overt phenotype within the first postnatal weeks. However, already as young adults, they displayed progressive motoric impairments. Moreover, as these mice grew older, they exhibited signs of a progeroid phenotype and early death. Histological analysis in moribund SOD2ako mice revealed the presence of age-related brain alterations, neuroinflammation, neuronal damage and myelin impairment in brain and spinal cord. Additionally, transcriptome analysis of primary astrocytes revealed that SOD2 deletion triggered a hypometabolic state and promoted polarization toward A1-neurotoxic status, possibly underlying the neuronal and myelin deficits. Conclusively, our study identifies maintenance of ROS homeostasis in astrocytes as a critical prerequisite for physiological CNS aging.


Assuntos
Envelhecimento , Astrócitos , Doenças Neurodegenerativas , Animais , Camundongos , Sistema Nervoso Central , Oxirredução
4.
Cell Mol Gastroenterol Hepatol ; 16(5): 783-807, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37543088

RESUMO

BACKGROUND AND AIMS: Development of pancreatic ductal adenocarcinoma (PDAC) is a multistep process intensively studied; however, precocious diagnosis and effective therapy still remain unsatisfactory. The role for Notch signaling in PDAC has been discussed controversially, as both cancer-promoting and cancer-antagonizing functions have been described. Thus, an improved understanding of the underlying molecular mechanisms is necessary. Here, we focused on RBPJ, the receiving transcription factor in the Notch pathway, examined its expression pattern in PDAC, and characterized its function in mouse models of pancreatic cancer development and in the regeneration process after acute pancreatitis. METHODS: Conditional transgenic mouse models were used for functional analysis of RBPJ in the adult pancreas, initiation of PDAC precursor lesions, and pancreatic regeneration. Pancreata and primary acinar cells were tested for acinar-to-ductal metaplasia together with immunohistology and comprehensive transcriptional profiling by RNA sequencing. RESULTS: We identified reduced RBPJ expression in a subset of human PDAC specimens. Ptf1α-CreERT-driven depletion of RBPJ in transgenic mice revealed that its function is dispensable for the homeostasis and maintenance of adult acinar cells. However, primary RBPJ-deficient acinar cells underwent acinar-to-ductal differentiation in ex vivo. Importantly, oncogenic KRAS expression in the context of RBPJ deficiency facilitated the development of pancreatic intraepithelial neoplasia lesions with massive fibrotic stroma formation. Interestingly, RNA-sequencing data revealed a transcriptional profile associated with the cytokine/chemokine and extracellular matrix changes. In addition, lack of RBPJ delays the course of acute pancreatitis and critically impairs it in the context of KRASG12D expression. CONCLUSIONS: Our findings imply that downregulation of RBPJ in PDAC patients derepresses Notch targets and promotes KRAS-mediated pancreatic acinar cells transformation and desmoplasia development.


Assuntos
Carcinoma in Situ , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Pancreatite , Animais , Humanos , Camundongos , Células Acinares/metabolismo , Doença Aguda , Carcinoma in Situ/metabolismo , Carcinoma Ductal Pancreático/patologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos Transgênicos , Neoplasias Pancreáticas/patologia , Pancreatite/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Neoplasias Pancreáticas
5.
Mol Neurodegener ; 18(1): 24, 2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37069623

RESUMO

BACKGROUND: Inflammaging represents an accepted concept where the immune system shifts to a low-grade chronic pro-inflammatory state without overt infection upon aging. In the CNS, inflammaging is mainly driven by glia cells and associated with neurodegenerative processes. White matter degeneration (WMD), a well-known process in the aging brain, manifests in myelin loss finally resulting in motor, sensory and cognitive impairments. Oligodendrocytes (OL) are responsible for homeostasis and maintenance of the myelin sheaths, which is a complex and highly energy demanding process sensitizing these cells to metabolic, oxidative and other forms of stress. Yet, the immediate impact of chronic inflammatory stress like inflammaging on OL homeostasis, myelin maintenance and WMD remains open. METHODS: To functionally analyze the role of IKK/NF-κB signaling in the regulation of myelin homeostasis and maintenance in the adult CNS, we established a conditional mouse model allowing NF-κB activation in mature myelinating oligodendrocytes. IKK2-CAPLP-CreERT2 mice were characterized by biochemical, immunohistochemical, ultrastructural and behavioral analyses. Transcriptome data from isolated, primary OLs and microglia cells were explored by in silico pathway analysis and validated by complementary molecular approaches. RESULTS: Chronic NF-κB activation in mature OLs leads to aggravated neuroinflammatory conditions phenocopying brain inflammaging. As a consequence, IKK2-CAPLP-CreERT2 mice showed specific neurological deficits and impaired motoric learning. Upon aging, persistent NF-κB signaling promotes WMD in these mice as ultrastructural analysis revealed myelination deficits in the corpus callosum accompanied by impaired myelin protein expression. RNA-Seq analysis of primary oligodendrocytes and microglia cells uncovers gene expression signatures associated with activated stress responses and increased post mitotic cellular senescence (PoMiCS) which was confirmed by elevated senescence-associated ß-galactosidase activity and SASP gene expression profile. We identified an elevated integrated stress response (ISR) characterized by phosphorylation of eIF2α as a relevant molecular mechanism which is able to affect translation of myelin proteins. CONCLUSIONS: Our findings demonstrate an essential role of IKK/NF-κB signaling in mature, post-mitotic OLs in regulating stress-induced senescence in these cells. Moreover, our study identifies PoMICS as an important driving force of age-dependent WMD as well as of traumatic brain injury induced myelin defects.


Assuntos
NF-kappa B , Substância Branca , Camundongos , Animais , NF-kappa B/metabolismo , Substância Branca/metabolismo , Oligodendroglia , Bainha de Mielina , Transdução de Sinais/fisiologia
6.
Sci Immunol ; 8(79): eade7953, 2023 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-36662884

RESUMO

Interferon regulatory factor 4 (IRF4) is a transcription factor (TF) and key regulator of immune cell development and function. We report a recurrent heterozygous mutation in IRF4, p.T95R, causing an autosomal dominant combined immunodeficiency (CID) in seven patients from six unrelated families. The patients exhibited profound susceptibility to opportunistic infections, notably Pneumocystis jirovecii, and presented with agammaglobulinemia. Patients' B cells showed impaired maturation, decreased immunoglobulin isotype switching, and defective plasma cell differentiation, whereas their T cells contained reduced TH17 and TFH populations and exhibited decreased cytokine production. A knock-in mouse model of heterozygous T95R showed a severe defect in antibody production both at the steady state and after immunization with different types of antigens, consistent with the CID observed in these patients. The IRF4T95R variant maps to the TF's DNA binding domain, alters its canonical DNA binding specificities, and results in a simultaneous multimorphic combination of loss, gain, and new functions for IRF4. IRF4T95R behaved as a gain-of-function hypermorph by binding to DNA with higher affinity than IRF4WT. Despite this increased affinity for DNA, the transcriptional activity on IRF4 canonical genes was reduced, showcasing a hypomorphic activity of IRF4T95R. Simultaneously, IRF4T95R functions as a neomorph by binding to noncanonical DNA sites to alter the gene expression profile, including the transcription of genes exclusively induced by IRF4T95R but not by IRF4WT. This previously undescribed multimorphic IRF4 pathophysiology disrupts normal lymphocyte biology, causing human disease.


Assuntos
Regulação da Expressão Gênica , Fatores Reguladores de Interferon , Camundongos , Animais , Humanos , Linfócitos B , DNA/metabolismo , Mutação
7.
Brain Behav Immun ; 108: 148-161, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36427809

RESUMO

Stress-associated somatic and psychiatric disorders are often linked to non-resolving low-grade inflammation, which is promoted at least in part by glucocorticoid (GC) resistance of distinct immune cell subpopulations. While the monocyte/macrophage compartment was in the focus of many clinical and preclinical studies, the role of myeloid-derived suppressor cells (MDSCs) in stress-associated pathologies and GC resistance is less understood. As GC resistance is a clear risk factor for posttraumatic complications in patients on intensive care, the exact interplay of physical and psychosocial traumatization in the development of GC resistance needs to be further clarified. In the current study we employ the chronic subordinate colony housing (CSC) paradigm, a well-characterized mouse model of chronic psychosocial stress, to study the role of myeloid cells, in particular of MDSCs, in innate immune activation and GC resistance following combined psychosocial and physical (e.g., bite wounds) trauma. Our findings support the hypothesis that stress-induced neutrophils, polymorphonuclear (PMN)-MDSCs and monocytes/monocyte-like (MO)-MDSCs get primed and activated locally in the bone marrow as determined by toll-like receptor (TLR)2 upregulation and increased basal and lipopolysaccharide (LPS)-induced in vitro cell viability. These primed and activated myeloid cells emigrate into the peripheral circulation and subsequently, if CSC is accompanied by significant bite wounding, accumulate in the spleen. Here, PMN-MDSCs and monocytes/MO-MDSCs upregulate TLR4 expression, which exclusively in PMN-MDSCs promotes NF-κB hyperactivation upon LPS-stimulation, thereby exceeding the anti-inflammatory capacities of GCs and resulting in GC resistance.


Assuntos
Glucocorticoides , Células Supressoras Mieloides , Estresse Psicológico , Animais , Camundongos , Glucocorticoides/farmacologia , Lipopolissacarídeos , Monócitos , Células Mieloides , Células Supressoras Mieloides/metabolismo
8.
Cancers (Basel) ; 13(19)2021 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-34638511

RESUMO

The Notch signaling pathway is an evolutionary conserved signal transduction cascade present in almost all tissues and is required for embryonic and postnatal development, as well as for stem cell maintenance, but it is also implicated in tumorigenesis including pancreatic cancer and leukemia. The transcription factor RBPJ forms a coactivator complex in the presence of a Notch signal, whereas it represses Notch target genes in the absence of a Notch stimulus. In the pancreas, a specific paralog of RBPJ, called RBPJL, is expressed and found as part of the heterotrimeric PTF1-complex. However, the function of RBPJL in Notch signaling remains elusive. Using molecular modeling, biochemical and functional assays, as well as single-molecule time-lapse imaging, we show that RBPJL and RBPJ, despite limited sequence homology, possess a high degree of structural similarity. RBPJL is specifically expressed in the exocrine pancreas, whereas it is mostly undetectable in pancreatic tumour cell lines. Importantly, RBPJL is not able to interact with Notch-1 to -4 and it does not support Notch-mediated transactivation. However, RBPJL can bind to canonical RBPJ DNA elements and shows migration dynamics comparable to that of RBPJ in the nuclei of living cells. Importantly, RBPJL is able to interact with SHARP/SPEN, the central corepressor of the Notch pathway. In line with this, RBPJL is able to fully reconstitute transcriptional repression at Notch target genes in cells lacking RBPJ. Together, RBPJL can act as an antagonist of RBPJ, which renders cells unresponsive to the activation of Notch.

9.
Cells ; 10(10)2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34685649

RESUMO

Alzheimer's disease (AD) is a common neurodegenerative disease that is accompanied by pronounced neuroinflammatory responses mainly characterized by marked microgliosis and astrogliosis. However, it remains open as to how different aspects of astrocytic and microglial activation affect disease progression. Previously, we found that microglia expansion in the spinal cord, initiated by IKK2/NF-κB activation in astrocytes, exhibits stage-dependent beneficial effects on the progression of amyotrophic lateral sclerosis. Here, we investigated the impact of NF-κB-initiated neuroinflammation on AD pathogenesis using the APP23 mouse model of AD in combination with conditional activation of IKK2/NF-κB signaling in astrocytes. We show that NF-κB activation in astrocytes triggers a distinct neuroinflammatory response characterized by striking astrogliosis as well as prominent microglial reactivity. Immunohistochemistry and Congo red staining revealed an overall reduction in the size and number of amyloid plaques in the cerebral cortex and hippocampus. Interestingly, isolated primary astrocytes and microglia cells exhibit specific marker gene profiles which, in the case of microglia, point to an enhanced plaque clearance capacity. In contrast, direct IKK2/NF-κB activation in microglia results in a pro-inflammatory polarization program. Our findings suggest that IKK2/NF-κB signaling in astrocytes may activate paracrine mechanisms acting on microglia function but also on APP processing in neurons.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Polaridade Celular , Quinase I-kappa B/metabolismo , Microglia/patologia , NF-kappa B/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Inflamação/patologia , Camundongos Transgênicos , Microglia/metabolismo , Modelos Biológicos , Fagocitose , Fenótipo , Placa Amiloide/genética , Placa Amiloide/patologia , Proteólise , Transdução de Sinais
10.
Cell Mol Life Sci ; 78(6): 2987-3003, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33206203

RESUMO

The pathogenesis of obesity-related metabolic diseases has been linked to the inflammation of white adipose tissue (WAT), but the molecular interconnections are still not fully understood. MiR-146a controls inflammatory processes by suppressing pro-inflammatory signaling pathways. The aim of this study was to characterize the role of miR-146a in obesity and insulin resistance. MiR-146a-/- mice were subjected to a high-fat diet followed by metabolic tests and WAT transcriptomics. Gain- and loss-of-function studies were performed using human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes. Compared to controls, miR-146a-/- mice gained significantly more body weight on a high-fat diet with increased fat mass and adipocyte hypertrophy. This was accompanied by exacerbated liver steatosis, insulin resistance, and glucose intolerance. Likewise, adipocytes transfected with an inhibitor of miR-146a displayed a decrease in insulin-stimulated glucose uptake, while transfecting miR-146a mimics caused the opposite effect. Natriuretic peptide receptor 3 (NPR3) was identified as a direct target gene of miR-146a in adipocytes and CRISPR/Cas9-mediated knockout of NPR3 increased insulin-stimulated glucose uptake and enhanced de novo lipogenesis. In summary, miR-146a regulates systemic and adipocyte insulin sensitivity via downregulation of NPR3.


Assuntos
Resistência à Insulina , MicroRNAs/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Adipócitos/citologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Animais , Antagomirs/metabolismo , Peso Corporal , Dieta Hiperlipídica , Fígado Gorduroso/patologia , Teste de Tolerância a Glucose , Humanos , Resistência à Insulina/genética , Lipogênese , Fígado/metabolismo , Camundongos , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo , Triglicerídeos/metabolismo
11.
Life Sci Alliance ; 3(11)2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32900826

RESUMO

Blood-spinal cord barrier (BSCB) disruption is thought to contribute to motoneuron (MN) loss in amyotrophic lateral sclerosis (ALS). It is currently unclear whether impairment of the BSCB is the cause or consequence of MN dysfunction and whether its restoration may be directly beneficial. We revealed that SOD1 G93A , FUS ΔNLS , TDP43 G298S , and Tbk1 +/- ALS mouse models commonly shared alterations in the BSCB, unrelated to motoneuron loss. We exploit PSAM/PSEM chemogenetics in SOD1 G93A mice to demonstrate that the BSCB is rescued by increased MN firing, whereas inactivation worsens it. Moreover, we use DREADD chemogenetics, alone or in multiplexed form, to show that activation of Gi signaling in astrocytes restores BSCB integrity, independently of MN firing, with no effect on MN disease markers and dissociating them from BSCB disruption. We show that astrocytic levels of the BSCB stabilizers Wnt7a and Wnt5a are decreased in SOD1 G93A mice and strongly enhanced by Gi signaling, although further decreased by MN inactivation. Thus, we demonstrate that BSCB impairment follows MN dysfunction in ALS pathogenesis but can be reversed by Gi-induced expression of astrocytic Wnt5a/7a.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Astrócitos/metabolismo , Neurônios Motores/metabolismo , Esclerose Lateral Amiotrófica/sangue , Animais , Astrócitos/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/fisiologia , Medula Espinal/metabolismo , Coluna Vertebral/irrigação sanguínea , Coluna Vertebral/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt-5a/metabolismo
12.
Photoacoustics ; 18: 100161, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32021797

RESUMO

Photoacoustic spectroscopy employs acoustic resonators for signal amplification. Resonators are usually closed, however, in some applications, open resonators are preferred. The opening deteriorates the photoacoustic signal hence reducing the sensitivity of the photoacoustic measurement. We present two new approaches for simulating the photoacoustic signal in open resonators using finite element modelling. The approaches are based on the amplitude mode expansion model and the viscothermal model with the opening modelled using perfectly matched layers and the boundary element method respectively. Additionally, the performance of the viscothermal model using perfectly matched layers for simulating open resonators is extended to the ultrasound region. The simulation results are verified by comparing them to photoacoustic measurements. The approaches provide an accurate basis for designing and optimizing open resonators with high sensitivity.

13.
J Endocrinol ; 244(2): 323-337, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31682591

RESUMO

Maturity-onset diabetes of the young (MODY) is a group of monogenetic forms of diabetes mellitus caused by mutations in genes regulating ß-cell development and function. MODY represents a heterogeneous group of non-insulin-dependent diabetes arising in childhood or adult life. Interestingly, clinical heterogeneity in MODY patients like variable disease onset and severity is observed even among individual family members sharing the same mutation, an issue that is not well understood. As high blood glucose levels are a well-known factor promoting ß-cell stress and ultimately leading to cell death, we asked whether additional ß-cell stress might account for the occurrence of disease heterogeneity in mice carrying a MODY4 mutation. In order to challenge ß-cells, we established a MODY4 animal model based on Pdx1 (pancreatic and duodenal homeobox 1) haploinsufficiency, which allows conditional modulation of cell stress by genetic inhibition of the stress-responsive IKK/NF-κB signalling pathway. While Pdx1+/- mice were found glucose intolerant without progressing to diabetes, additional challenge of ß-cell function by IKK/NF-κB inhibition promoted rapid diabetes development showing hyperglycaemia, hypoinsulinemia and loss of ß-cell mass. Disease pathogenesis was characterized by deregulation of genes controlling ß-cell homeostasis and function. Importantly, restoration of normal IKK/NF-κB signalling reverted the diabetic phenotype including normalization of glycaemia and ß-cell mass. Our findings implicate that the avoidance of additional ß-cell stress can delay a detrimental disease progression in MODY4 diabetes. Remarkably, an already present diabetic phenotype can be reversed when ß-cell stress is normalized.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Glicemia/metabolismo , Morte Celular , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Estresse Fisiológico , Transativadores/genética , Transativadores/metabolismo
14.
Cell Rep ; 29(9): 2862-2874.e9, 2019 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-31775051

RESUMO

Intracellular accumulation of α-synuclein (α-syn) and formation of Lewy bodies are neuropathological characteristics of Parkinson's disease (PD) and related α-synucleinopathies. Oligomerization and spreading of α-syn from neuron to neuron have been suggested as key events contributing to the progression of PD. To directly visualize and characterize α-syn oligomerization and spreading in vivo, we generated two independent conditional transgenic mouse models based on α-syn protein complementation assays using neuron-specifically expressed split Gaussia luciferase or split Venus yellow fluorescent protein (YFP). These transgenic mice allow direct assessment of the quantity and subcellular distribution of α-syn oligomers in vivo. Using these mouse models, we demonstrate an age-dependent accumulation of a specific subtype of α-syn oligomers. We provide in vivo evidence that, although α-syn is found throughout neurons, α-syn oligomerization takes place at the presynapse. Furthermore, our mouse models provide strong evidence for a transsynaptic cell-to-cell transfer of de novo generated α-syn oligomers in vivo.


Assuntos
Neurônios/metabolismo , Doença de Parkinson/genética , alfa-Sinucleína/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos
15.
Cancer Cell ; 36(3): 250-267.e9, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31526758

RESUMO

How lymphoma cells (LCs) invade the brain during the development of central nervous system lymphoma (CNSL) is unclear. We found that NF-κB-induced gliosis promotes CNSL in immunocompetent mice. Gliosis elevated cell-adhesion molecules, which increased LCs in the brain but was insufficient to induce CNSL. Astrocyte-derived CCL19 was required for gliosis-induced CNSL. Deleting CCL19 in mice or CCR7 from LCs abrogated CNSL development. Two-photon microscopy revealed LCs transiently entering normal brain parenchyma. Astrocytic CCL19 enhanced parenchymal CNS retention of LCs, thereby promoting CNSL formation. Aged, gliotic wild-type mice were more susceptible to forming CNSL than young wild-type mice, and astrocytic CCL19 was observed in both human gliosis and CNSL. Therefore, CCL19-CCR7 interactions may underlie an increased age-related risk for CNSL.


Assuntos
Envelhecimento/patologia , Neoplasias do Sistema Nervoso Central/patologia , Quimiocina CCL19/metabolismo , Gliose/patologia , Linfoma/patologia , Adolescente , Adulto , Idoso , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/diagnóstico por imagem , Barreira Hematoencefálica/patologia , Linhagem Celular Tumoral/transplante , Neoplasias do Sistema Nervoso Central/diagnóstico por imagem , Neoplasias do Sistema Nervoso Central/cirurgia , Quimiocina CCL19/genética , Quimiocina CXCL12 , Modelos Animais de Doenças , Feminino , Gliose/diagnóstico por imagem , Humanos , Microscopia Intravital , Linfoma/diagnóstico por imagem , Linfoma/cirurgia , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Receptores CCR7/genética , Receptores CCR7/metabolismo , Imagem com Lapso de Tempo , Adulto Jovem
16.
Sensors (Basel) ; 19(13)2019 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-31261919

RESUMO

T-cell resonators have been used lately for non-invasive blood glucose measurements for photoacoustic spectroscopy on skin samples. A resonator has a significant role in determining the strength of the measured signal and the overall sensitivity of the sensor. Here we present results of the measurement of the photoacoustic signal of such a T-cell resonator. The signal is also modelled using the amplitude mode expansion method, which is based on eigenmode expansion and the introduction of losses in the form of loss factors. The measurement reproduced almost all the calculated resonances from the numerical models with fairly good agreement. The cause of the differences between the measured and the simulated resonances are explained. In addition, the amplitude mode expansion simulation model is established as a faster and computationally less demanding photoacoustic simulation alternative to the viscothermal model. The resonance frequencies from the two models differ by less than 1.8%. It is noted that the relative height of the amplitudes from the two models depends on the location of the antinodes within the different parts of the resonator. The amplitude mode expansion model provides a quick simulation tool for the optimization and design of macro resonators.


Assuntos
Técnicas Biossensoriais , Glucose/isolamento & purificação , Técnicas Fotoacústicas , Linfócitos T/metabolismo , Simulação por Computador , Glucose/metabolismo , Humanos , Sistemas de Infusão de Insulina , Linfócitos T/química
17.
Brain Behav Immun ; 81: 228-246, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31207335

RESUMO

Traumatic brain injury (TBI) and ethanol intoxication (EI) frequently coincide, particularly in young subjects. However, the mechanisms of their interaction remain poorly understood. Among other pathogenic pathways, TBI induces glial activation and neuroinflammation in the hippocampus, resulting in acute and chronic hippocampal dysfunction. In this regard, we investigated the role of EI affecting these responses unfolding after TBI. We used a blunt, weight-drop approach to model TBI in mice. Male mice were pre-administered with ethanol or vehicle to simulate EI. The neuroinflammatory response in the hippocampus was assessed by monitoring the expression levels of >20 cytokines, the phosphorylation status of transcription factors and the phenotype of microglia and astrocytes. We used AS1517499, a brain-permeable STAT6 inhibitor, to elucidate the role of this pathway in the EI/TBI interaction. We showed that TBI causes the elevation of IL-33, IL-1ß, IL-38, TNF-α, IFN-α, IL-19 in the hippocampus at 3 h time point and concomitant EI results in the dose-dependent downregulation of IL-33, IL-1ß, IL-38, TNF-α and IL-19 (but not of IFN-α) and in the selective upregulation of IL-13 and IL-12. EI is associated with the phosphorylation of STAT6 and the transcription of STAT6-controlled genes. Moreover, ethanol-induced STAT6 phosphorylation and transcriptional activation can be recapitulated in vitro by concomitant exposure of neurons to ethanol, depolarization and inflammatory stimuli (simulating the acute trauma). Acute STAT6 inhibition prevents the effects of EI on IL-33 and TNF-α, but not on IL-13 and negates acute EI beneficial effects on TBI-associated neurological impairment. Additionally, EI is associated with reduced microglial activation and astrogliosis as well as preserved synaptic density and baseline neuronal activity 7 days after TBI and all these effects are prevented by acute administration of the STAT6 inhibitor concomitant to EI. EI concomitant to TBI exerts significant immunomodulatory effects on cytokine induction and microglial activation, largely through the activation of STAT6 pathway, ultimately with beneficial outcomes.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Etanol/farmacologia , Fator de Transcrição STAT6/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação de Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Microglia/metabolismo , Microglia/patologia , Neuroimunomodulação/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fator de Transcrição STAT6/imunologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
18.
Proc Natl Acad Sci U S A ; 116(3): 970-975, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30591564

RESUMO

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a critical regulator of cell death and inflammation, but its relevance for human disease pathogenesis remains elusive. Studies of monogenic disorders might provide critical insights into disease mechanisms and therapeutic targeting of RIPK1 for common diseases. Here, we report on eight patients from six unrelated pedigrees with biallelic loss-of-function mutations in RIPK1 presenting with primary immunodeficiency and/or intestinal inflammation. Mutations in RIPK1 were associated with reduced NF-κB activity, defective differentiation of T and B cells, increased inflammasome activity, and impaired response to TNFR1-mediated cell death in intestinal epithelial cells. The characterization of RIPK1-deficient patients highlights the essential role of RIPK1 in controlling human immune and intestinal homeostasis, and might have critical implications for therapies targeting RIPK1.


Assuntos
Diferenciação Celular , Imunidade nas Mucosas/genética , Doenças Inflamatórias Intestinais , Mucosa Intestinal , Proteína Serina-Treonina Quinases de Interação com Receptores , Imunodeficiência Combinada Severa , Linfócitos B/imunologia , Linfócitos B/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Células HCT116 , Células HEK293 , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Mutação , NF-kappa B/genética , NF-kappa B/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Linfócitos T/imunologia , Linfócitos T/patologia
19.
EMBO J ; 37(16)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29875132

RESUMO

Astrocytes are involved in non-cell-autonomous pathogenic cascades in amyotrophic lateral sclerosis (ALS); however, their role is still debated. We show that astrocytic NF-κB activation drives microglial proliferation and leukocyte infiltration in the SOD1 (G93A) ALS model. This response prolongs the presymptomatic phase, delaying muscle denervation and decreasing disease burden, but turns detrimental in the symptomatic phase, accelerating disease progression. The transition corresponds to a shift in the microglial phenotype showing two effects that can be dissociated by temporally controlling NF-κB activation. While NF-κB activation in astrocytes induced a Wnt-dependent microglial proliferation in the presymptomatic phase with neuroprotective effects on motoneurons, in later stage, astrocyte NF-κB-dependent microglial activation caused an accelerated disease progression. Notably, suppression of the early microglial response by CB2R agonists had acute detrimental effects. These data identify astrocytes as important regulators of microglia expansion and immune response. Therefore, stage-dependent microglia modulation may be an effective therapeutic strategy in ALS.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Astrócitos/imunologia , NF-kappa B/imunologia , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/terapia , Animais , Astrócitos/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Microglia/imunologia , Microglia/patologia , Neurônios Motores/imunologia , Neurônios Motores/patologia , NF-kappa B/genética , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/imunologia , Superóxido Dismutase/genética , Superóxido Dismutase/imunologia , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/imunologia
20.
Sci Rep ; 8(1): 5527, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29615749

RESUMO

Due to the highly invasive nature of Glioblastoma (GB), complete surgical resection is not feasible, while motile tumour cells are often associated with several specific brain structures that enhance treatment-resistance. Here, we investigate the therapeutic potential of Disulfiram and Carbenoxolone, that inhibit two distinct interactions between GB and the brain tissue microenvironment: stress-induced cell-matrix adhesion and gap junction mediated cell-cell communication, respectively. Increase in cell numbers of tumour-initiating cells, which are cultured in suspension as cell clusters, and adherent differentiated cells can be blocked to a similar extent by Carbenoxolone, as both cell populations form gap junctions, but the adherent differentiated cells are much more sensitive to Disulfiram treatment, which - via modulation of NF-κB signalling - interferes with cell-substrate adhesion. Interestingly, inducing adhesion in tumour-initiating cells without differentiating them does not sensitize for Disulfiram. Importantly, combining Disulfiram, Carbenoxolone and the standard chemotherapeutic drug Temozolomide reduces tumour size in an orthotopic mouse model. Isolating GB cells from their direct environment within the brain represents an important addition to current therapeutic approaches. The blockage of cellular interactions via the clinically relevant substances Disulfiram and Carbenoxolone, has distinct effects on different cell populations within a tumour, potentially reducing motility and/or resistance to apoptosis.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Carbenoxolona/farmacologia , Dissulfiram/farmacologia , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Inibidores de Acetaldeído Desidrogenases/farmacologia , Animais , Antiulcerosos/farmacologia , Apoptose , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Adesão Celular , Proliferação de Células , Quimioterapia Combinada , Perfilação da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Prognóstico , Transdução de Sinais , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...