Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Neurosci ; 43(2): 319-332, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36446585

RESUMO

Mechanical impact-induced primary injury after traumatic brain injury (TBI) leads to acute microglial pro-inflammatory activation and consequently mediates neurodegeneration, which is a major secondary brain injury mechanism. However, the detailed pathologic cascades have not been fully elucidated, partially because of the pathologic complexity in animal TBI models. Although there are several in vitro TBI models, none of them closely mimic post-TBI microglial activation. In the present study, we aimed to establish an in vitro TBI model, specifically reconstituting the pro-inflammatory activation and associated neurodegeneration following TBI. We proposed three sets of experiments. First, we established a needle scratch injured neuron-induced microglial activation and neurodegeneration in vitro model of TBI. Second, we compared microglial pro-inflammatory cytokines profiles between the in vitro TBI model and TBI in male mice. Additionally, we validated the role of injured neurons-derived damage-associated molecular patterns in amplifying microglial pro-inflammatory pathways using the in vitro TBI model. Third, we applied the in vitro model for the first time to characterize the cellular metabolic profile of needle scratch injured-neuron-activated microglia, and define the role of metabolic reprogramming in mediating pro-inflammatory microglial activation and mediated neurodegeneration. Our results showed that we successfully established a novel in vitro TBI model, which closely mimics primary neuronal injury-triggered microglial pro-inflammatory activation and mediated neurodegeneration after TBI. This in vitro model provides an advanced and highly translational platform for dissecting interactions in the pathologic processes of neuronal injury-microglial activation-neuronal degeneration cascade, and elucidating the detailed underlying cellular and molecular insights after TBI.SIGNIFICANCE STATEMENT Microglial activation is a key component of acute neuroinflammation that leads to neurodegeneration and long-term neurologic outcome deficits after TBI. However, it is not feasible to truly dissect primary neuronal injury-induced microglia activation, and consequently mediated neurodegeneration in vivo Furthermore, there is currently lacking of in vitro TBI models closely mimicking the TBI primary injury-mediated microglial activation. In this study, we successfully established and validated a novel in vitro TBI model of microglial activation, and for the first time, characterized the cellular metabolic profile of microglia in this model. This novel microglial activation in vitro TBI model will help in elucidating microglial inflammatory activation and consequently associated neurodegeneration after TBI.


Assuntos
Lesões Encefálicas Traumáticas , Microglia , Camundongos , Masculino , Animais , Microglia/metabolismo , Lesões Encefálicas Traumáticas/patologia , Macrófagos/metabolismo , Neurônios/metabolismo , Camundongos Endogâmicos C57BL
2.
Ann Hematol ; 103(5): 1423-1433, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-37405444

RESUMO

Coronavirus disease 19 (COVID-19) is an infectious disease caused by severe acute respiratory coronavirus 2 (SARS-CoV-2) causing acute systemic disorders and multi-organ damage. ß-thalassemia (ß-T) is an autosomal recessive disorder leading to the development of anemia. ß-T may lead to complications such as immunological disorders, iron overload, oxidative stress, and endocrinopathy. ß-T and associated complications may increase the risk of SARS-CoV-2, as inflammatory disturbances and oxidative stress disorders are linked with COVID-19. Therefore, the objective of the present review was to elucidate the potential link between ß-T and COVID-19 regarding the underlying comorbidities. The present review showed that most of the ß-T patients with COVID-19 revealed mild to moderate clinical features, and ß-T may not be linked with Covid-19 severity. Though patients with transfusion-dependent ß-T (TDT) develop less COVID-19 severity compared to non-transfusion-depend ß-T(NTDT), preclinical and clinical studies are recommended in this regard.


Assuntos
COVID-19 , Sobrecarga de Ferro , Talassemia beta , Humanos , Talassemia beta/complicações , Talassemia beta/epidemiologia , Talassemia beta/terapia , COVID-19/complicações , SARS-CoV-2 , Transfusão de Sangue , Sobrecarga de Ferro/etiologia
3.
Small ; 19(27): e2207433, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36978239

RESUMO

The intracellular uptake and interaction behavior of emulsion microparticles in liver cells critical to host defense and inflammation is significant to understanding their potential cytotoxicity and biomedical applications. In this study, the cell death responses of fibroblastic, hepatocyte, and Kupffer cells (KCs) induced by four types of emulsion particles that are stabilized by polysaccharide nanofibers (cellulose or chitin), an inorganic nanoparticle (ß-tricalcium phosphate), or surfactants are compared. Pickering emulsion (PE) microparticles stabilized by polysaccharide nanofibers or inorganic nanoparticles have a droplet size of 1-3 µm, while the surfactant-stabilized emulsion has a diameter of ≈190 nm. Polysaccharide nanofiber-stabilized PEs (PPEs) markedly induce lactate dehydrogenase release in all cell types. Additionally, characteristic pyroptotic cell death, which is accompanied by cell swelling, membrane blebbing, and caspase-1 activation, occurs in hepatocytes and KCs. These PE microparticles are co-cultured with lipopolysaccharide-primed KCs associated with cytokine interleukin-1ß release, and the PPEs demonstrate biological activity as a mediator of the inflammation response. Well-designed PPE microparticles induce pyroptosis of liver cells, which may provide new insight into regulating inflammation-related diseases for designing potent anticancer drugs and vaccine adjuvants.


Assuntos
Células de Kupffer , Nanofibras , Humanos , Células de Kupffer/metabolismo , Piroptose , Emulsões , Hepatócitos/metabolismo , Celulose , Inflamação/metabolismo
4.
Plant Foods Hum Nutr ; 78(1): 132-138, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36370293

RESUMO

Yarrow (Achillea millefolium L., AM) and nettle (Urtica dioica L., UD) are bioactive plants used commercially in functional food and supplement applications and traditionally to alleviate gastric disorders. In this work, the effects of food-grade optimized extracts of Finnish early-season AM and UD were tested on bacterial growth including potential beneficial and foodborne pathogens, as well as murine norovirus (MNV). The anti-inflammatory properties of the extracts were also tested in vitro by NF-κB reporter cells. The food-grade extraction was optimized with the response surface modelling in terms of total carotenoid, chlorophyll, and phenolic compounds contents and antioxidant capacities. The optimal food-grade extraction parameters were a 1-h extraction in 70% ethanol at 45 °C for AM, and at 49 °C for UD. There were no significant effects on the beneficial bacteria (Lacticaseibacillus and Bifidobacterium strains), and the extracts were more effective against gram-positive than gram-negative foodborne bacteria and potential pathogens. Listeria innocua was the most susceptible strain in the optimized extracts with a growth rate of 0.059 ± 0.004 for AM and 0.067 ± 0.006 for UD, p < 0.05 compared to control. The optimized extracts showed a logarithmic growth reduction of 0.67 compared to MNV. The hydroethanolic extracts were cytotoxic to both cell lines, whereas aqueous AM and UD extracts induced and reduced TLR4 signalling in a reporter cell line, respectively. The results provide novel food-grade extraction parameters and support the bioactive effects of AM and UD in functional food applications, but more research is needed to elucidate the precise biological activity in vivo for gastric health.


Assuntos
Achillea , Urtica dioica , Camundongos , Animais , Extratos Vegetais/farmacologia , Folhas de Planta , Antioxidantes/farmacologia , Bactérias
5.
Cell Mol Life Sci ; 78(4): 1637-1653, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32725371

RESUMO

Polysialic acid (polySia) emerges as a novel regulator of microglia activity. We recently identified polysialylated proteins in the Golgi compartment of murine microglia that are released in response to inflammatory stimulation. Since exogenously added polySia is able to attenuate the inflammatory response, we proposed that the release of polysialylated proteins constitutes a mechanism for negative feedback regulation of microglia activation. Here, we demonstrate that translocation of polySia from the Golgi to the cell surface can be induced by calcium depletion of the Golgi compartment and that polysialylated proteins are continuously released for at least 24 h after the onset of inflammatory stimulation. The latter was unexpected, because polySia signals detected by immunocytochemistry are rapidly depleted. However, it indicates that the amount of released polySia is much higher than anticipated based on immunostaining. This may be crucial for microglial responses during traumatic brain injury (TBI), as we detected polySia signals in activated microglia around a stab wound in the adult mouse brain. In BV2 microglia, the putative polySia receptor Siglec-E is internalized during lipopolysaccharide (LPS)-induced activation and in response to polySia exposure, indicating interaction. Correspondingly, CRISPR/Cas9-mediated Siglec-E knockout prevents inhibition of pro inflammatory activation by exogenously added polySia and leads to a strong increase of the LPS response. A comparable increase of LPS-induced activation has been observed in microglia with abolished polySia synthesis. Together, these results indicate that the release of the microglia-intrinsic polySia pool, as implicated in TBI, inhibits the inflammatory response by acting as a trans-activating ligand of Siglec-E.


Assuntos
Inflamação/genética , Microglia/metabolismo , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/genética , Ácidos Siálicos/genética , Animais , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos B/genética , Sistemas CRISPR-Cas/genética , Células Cultivadas , Retroalimentação Fisiológica/efeitos dos fármacos , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/genética , Humanos , Inflamação/imunologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Microglia/imunologia , Microglia/patologia , Fagocitose/efeitos dos fármacos , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Ácidos Siálicos/imunologia
6.
Glia ; 69(11): 2717-2738, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34329508

RESUMO

The ramified morphology of microglia and the dynamics of their membrane protrusions are essential for their functions in central nervous system development, homeostasis, and disease. Although their ability to change and control shape critically depends on the actin and actomyosin cytoskeleton, the underlying regulatory mechanisms remain largely unknown. In this study, we systematically analyzed the actomyosin cytoskeleton and regulators downstream of the small GTPase RhoA in the control of microglia shape and function. Our results reveal that (i) Myh9 controls cortical tension levels and affects microglia protrusion formation, (ii) cofilin-mediated maintenance of actin turnover regulates microglia protrusion extension, and (iii) Myh10 influences microglia inflammatory activation. Overall we uncover molecular pathways that regulate microglia morphology and identify type-II myosins as important regulators of microglia biology with differential roles in the control of cell shape (Myh9) and functions (Myh10).


Assuntos
Microglia , Miosinas , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Microglia/metabolismo , Miosinas/metabolismo
7.
J Cell Physiol ; 233(2): 1535-1547, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28600879

RESUMO

Multipotent mesenchymal stromal cells (MSCs) are considered cue regulators of tissue remodeling. Their activity is strongly governed by local milieu, where O2 level is most important. The elevation of inflammatory mediators and acute O2 lowering may additionally modulate MSC activity. In present paper the priming effects of IFN-gamma on adipose tissue-derived MSCs (ASCs) at tissue-related O2 level (5%) and acute hypoxic stress (0.1% O2 ) were assessed as alterations of ASCs' CFU-F, proliferation, migration, osteo-commitment. IFN-gamma priming provoked ROS elevation, cell growth slowdown, attenuation of both spontaneous and induced osteodifferentiation of tissue O2 -adapted ASCs. The prominent changes in ASC cytoskeleton-related gene transcription was detected. IFN-gamma exposure shifted the ASC paracrine profile, suppressing the production of VEGF and IL-8, while MCP-1 and IL-6 were stimulated. Conditioned medium of IFN-gamma-primed ASCs did not activate vessel growth in the CAM assay, but induced endothelial cell migration in "wound closure." Short-term hypoxia suppressed CFU-F number, IFN-gamma-induced elevation of IL-6 and endothelial cell migration, while it abolished IFN-gamma-provoked VEGF inhibition. After N-acetyl cysteine treatment ROS level was partly abolished providing additional enhancement of IL-6 and suppression of IL-8 and VEGF production. These findings demonstrated that paracrine activity of ASCs in part may be governed by ROS level. Thus, this study first demonstrated that IFN-gamma priming itself and in combination with acute O2 deprivation could supply dual effects on ASC functions providing both stimulatory and hampering effects. The equilibrium of these factors is a substantial requirement for the execution of MSC remodeling functions.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Interferon gama/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Hipóxia Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Coturnix , Meios de Cultivo Condicionados/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica , Osteogênese/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
8.
J Cell Biochem ; 119(3): 2875-2885, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29080356

RESUMO

Microgravity is a principal risk factor hampering human cardiovascular regulation during space flights. Endothelial dysfunction associated with the impaired integrity and uniformity of the monolayer represents a potential trigger for vascular damage. We characterized the expression profile of the multi-step cascade of adhesion molecules (ICAM-1, VCAM-1, E-selectin, VE-cadherin) in umbilical cord endothelial cells (ECs) after 24 h of exposure to simulated microgravity (SMG), pro-inflammatory cytokine TNF-α, and the combination of the two. Random Positioning Machine (RPM)-mediated SMG was used to mimic microgravity effects. SMG stimulated the expression of E-selectin, which is known to be involved in slowing leukocyte rolling. Primary ECs displayed heterogeneity with respect to the proportion of ICAM-1-positive cells. ECs were divided into two groups: pre-activated ECs displaying high proportion of ICAM-1+ -cells (ECs-1) (greater than 50%) and non-activated ECs with low proportion of ICAM-1+ -cells (ECs-2) (less than 25%). Only non-activated ECs-2 responded to SMG by elevating gene transcription and increasing ICAM-1 and VE-cadherin expression. This effect was enhanced after cumulative SMG-TNF-α exposure. ECs-1 displayed an unexpected decrease in number of E-selectin- and ICAM-1-positive ECs and pronounced up-regulation of VCAM1 upon activation of inflammation, which was partially abolished by SMG. Thus, non-activated ECs-2 are quite resistant to the impacts of microgravity and even exhibited an elevation of the VE-cadherin gene and protein expression, thus improving the integrity of the endothelial monolayer. Pre-activation of ECs with inflammatory stimuli may disturb the EC adhesion profile, attenuating its barrier function. These alterations may be among the mechanisms underlying cardiovascular dysregulation in real microgravity conditions.


Assuntos
Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Molécula 1 de Adesão Intercelular/biossíntese , Simulação de Ausência de Peso , Ausência de Peso , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos
9.
Circ Res ; 112(12): 1583-91, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23564640

RESUMO

RATIONALE: Hypoxia followed by reoxygenation promotes inflammation by activating nuclear factor κB transcription factors in endothelial cells (ECs). This process involves modification of the signaling intermediary tumor necrosis factor receptor-associated factor 6 with polyubiquitin chains. Thus, cellular mechanisms that suppress tumor necrosis factor receptor-associated factor 6 ubiquitination are potential therapeutic targets to reduce inflammation in hypoxic tissues. OBJECTIVE: In this study, we tested the hypothesis that endothelial activation in response to hypoxia-reoxygenation can be influenced by Cezanne, a deubiquitinating enzyme that cleaves ubiquitin from specific modified proteins. METHODS AND RESULTS: Studies of cultured ECs demonstrated that hypoxia (1% oxygen) induced Cezanne via p38 mitogen-activated protein kinase-dependent transcriptional and post-transcriptional mechanisms. Hypoxia-reoxygenation had minimal effects on proinflammatory signaling in unmanipulated ECs but significantly enhanced Lys63 polyubiquitination of tumor necrosis factor receptor-associated factor 6, activation of nuclear factor κB, and expression of inflammatory genes after silencing of Cezanne. Thus, although hypoxia primed cells for inflammatory activation, it simultaneously induced Cezanne, which impeded signaling to nuclear factor κB by suppressing tumor necrosis factor receptor-associated factor 6 ubiquitination. Similarly, ischemia induced Cezanne in the murine kidney in vascular ECs, glomerular ECs, podocytes, and epithelial cells, and genetic deletion of Cezanne enhanced renal inflammation and injury in murine kidneys exposed to ischemia followed by reperfusion. CONCLUSIONS: We conclude that inflammatory responses to ischemia are controlled by a balance between ubiquitination and deubiquitination, and that Cezanne is a key regulator of this process. Our observations have important implications for therapeutic targeting of inflammation and injury during ischemia-reperfusion.


Assuntos
Endopeptidases/metabolismo , Células Endoteliais/enzimologia , Inflamação/prevenção & controle , Rim/irrigação sanguínea , Traumatismo por Reperfusão/enzimologia , Fator 6 Associado a Receptor de TNF/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Modelos Animais de Doenças , Endopeptidases/deficiência , Endopeptidases/genética , Células Endoteliais/imunologia , Humanos , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Oxigênio/metabolismo , Interferência de RNA , Ratos , Ratos Endogâmicos F344 , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/genética , Fatores de Tempo , Transcrição Gênica , Transfecção , Ubiquitinação , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Clin Proteomics ; 11(1): 41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-26029019

RESUMO

BACKGROUND: Fibroblasts are mesenchymal stromal cells which occur in all tissue types. While their main function is related to ECM production and physical support, they are also important players in wound healing, and have further been recognized to be able to modulate inflammatory processes and support tumor growth. Fibroblasts can display distinct phenotypes, depending on their tissue origin, as well as on their functional state. RESULTS: In order to contribute to the proteomic characterization of fibroblasts, we have isolated primary human fibroblasts from human skin, lung and bone marrow and generated proteome profiles of these cells by LC-MS/MS. Comparative proteome profiling revealed characteristic differences therein, which seemed to be related to the cell's tissue origin. Furthermore, the cells were treated in vitro with the pro-inflammatory cytokine IL-1beta. While all fibroblasts induced the secretion of Interleukins IL-6 and IL-8 and the chemokine GRO-alpha, other inflammation-related proteins were up-regulated in an apparently tissue-dependent manner. Investigating fibroblasts from tumorous tissues of skin, lung and bone marrow with respect to such inflammation-related proteins revealed hardly any conformity but rather individual and tumor type-related variations. However, apparent up-regulation of IGF-II, PAI-1 and PLOD2 was observed in melanoma-, lung adenocarcinoma- and multiple myeloma-associated fibroblasts, as well as in hepatocellular carcinoma-associated fibroblasts. CONCLUSIONS: Inflammation-related proteome alterations of primary human fibroblasts were determined by the analysis of IL-1beta treated cells. Tumor-associated fibroblasts from different tissue types hardly showed signs of acute inflammation but displayed characteristic functional aberrations potentially related to chronic inflammation. The present data suggest that the state of the tumor microenvironment is relevant for tumor progression and targeted treatment of tumor-associated fibroblasts may support anti-cancer strategies.

11.
Curr Protoc ; 4(5): e1026, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38733265

RESUMO

Nuclear factor-κB (NF-κB) is a crucial pro-inflammatory transcription factor whose activation is of immense interest to immunology research. Estimation of NF-κB activation through flow cytometry is not possible due to the unavailability of robust flow cytometry antibodies that can bind to its phosphorylated, active, nuclear form. In this protocol, we describe a flow cytometry assay that measures the activation of the pro-inflammatory transcription factor NF-κB in stimulated immune cells by quantifying the degradation of its upstream regulator IκBα. We demonstrate the utility of this protocol by assessment of intracellular IκBα in human primary regulatory T cells experiencing TNFR2 agonism, a process previously reported to activate NF-κB in these cells. We also show that this assay may be applied to study NF-κB activation in other cell types, such as human primary T cells and THP-1 cell-derived macrophages, when induced by their corresponding inflammatory cues. Thus, this robust and reproducible protocol will be of interest to a wide range of scientists who aim to measure NF-κB activity in medium-to-high-throughput assays. © 2024 Wiley Periodicals LLC. Basic Protocol: Quantifying inflammatory activation by flow cytometry of IκBα degradation Support Protocol 1: Isolating and expanding human regulatory T cells Support Protocol 2: Calculating IC50 from flow cytometry data using Excel.


Assuntos
Citometria de Fluxo , Inibidor de NF-kappaB alfa , NF-kappa B , Humanos , Citometria de Fluxo/métodos , NF-kappa B/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Proteólise , Células THP-1 , Macrófagos/metabolismo , Macrófagos/imunologia
12.
J Nutr Biochem ; 115: 109281, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36758838

RESUMO

Transient high salt intake causes a sustained increase in blood pressure (BP) even after returning to a normal-salt diet, a phenomenon known as "salt memory." However, the molecular mechanisms of this phenomenon remain to be elucidated. Dahl salt-sensitive (SS) rats were fed a high-salt (8% NaCl) or high-salt diet and treated with drugs for 8 to 16 weeks and then returned to a normal-salt diet for 3 months. This study investigated the molecular mechanisms of salt memory and its mediation of SS hypertension and renal damage. We show that transient high salt intake caused persistent elevation of BP and exacerbation of kidney damage in Dahl SS rats even after returning to a normal-salt diet. Both epigenetic changes and inflammatory activation also persisted after resumption of a normal diet. Arterial BP, renal injury and the inflammatory response returned to normal levels in rats administered mycophenolate mofetil (MMF) during the 8-week period of high salt intake, resulting in the disappearance of salt memory. However, the vasodilator hydralazine did not ameliorate kidney damage or inflammatory activation, although it decreased BP to control levels. Transient high salt intake increased histone 3 lysine 4 monomethylation (H3K4me1) levels at the nuclear factor κB (NF-κB) subunit p65 promoter in SS rats, promoting p65 gene transcription and NF-κB activation and further leading to a series of inflammatory responses. Our findings demonstrate that transient high salt-induced epigenetic changes and persistent inflammatory activation play important roles in salt memory and its mediation of SS hypertension and renal damage.


Assuntos
Hipertensão , Nefropatias , Ratos , Animais , Cloreto de Sódio na Dieta/efeitos adversos , Cloreto de Sódio/efeitos adversos , NF-kappa B , Ratos Endogâmicos Dahl , Rim , Hipertensão/genética , Hipertensão/tratamento farmacológico , Pressão Sanguínea
13.
Int Immunopharmacol ; 121: 110491, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37329807

RESUMO

15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) exhibited potential to alleviate liver inflammation in chronic injury but was less studied in acute injury. Acute liver injury was associated with elevated macrophage migration inhibitory factor (MIF) levels in damaged hepatocytes. This study aimed to investigate the regulatory mechanism of hepatocyte-derived MIF by 15d-PGJ2 and its subsequent impact on acute liver injury. In vivo, mouse models were established by carbon tetrachloride (CCl4) intraperitoneal injection, with or without 15d-PGJ2 administration. 15d-PGJ2 treatment reduced the necrotic areas induced by CCl4. In the same mouse model constructed using enhanced green fluorescent protein (EGFP)-labeled bone marrow (BM) chimeric mice, 15d-PGJ2 reduced CCl4 induced BM-derived macrophage (BMM, EGFP+F4/80+) infiltration and inflammatory cytokine expression. Additionally, 15d-PGJ2 down-regulated liver and serum MIF levels; liver MIF expression was positively correlated with BMM percentage and inflammatory cytokine expression. In vitro, 15d-PGJ2 inhibited Mif expression in hepatocytes. In primary hepatocytes, reactive oxygen species inhibitor (NAC) showed no effect on MIF inhibition by 15d-PGJ2; PPARγ inhibitor (GW9662) abolished 15d-PGJ2 suppressed MIF expression and antagonists (troglitazone, ciglitazone) mimicked its function. In Pparg silenced AML12 cells, the suppression of MIF by 15d-PGJ2 was weakened; 15d-PGJ2 promoted PPARγ activation in AML 12 cells and primary hepatocytes. Furthermore, the conditioned medium of recombinant MIF- and lipopolysaccharide-treated AML12 respectively promoted BMM migration and inflammatory cytokine expression. Conditioned medium of 15d-PGJ2- or siMif-treated injured AML12 suppressed these effects. Collectively, 15d-PGJ2 activated PPARγ to suppress MIF expression in injured hepatocytes, reducing BMM infiltration and pro-inflammatory activation, ultimately alleviating acute liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Fatores Inibidores da Migração de Macrófagos , Prostaglandina D2 , Animais , Camundongos , Meios de Cultivo Condicionados , Hepatócitos , Fígado , Fatores Inibidores da Migração de Macrófagos/metabolismo , PPAR gama , Prostaglandina D2/uso terapêutico , Prostaglandina D2/farmacologia , Prostaglandinas , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico
14.
Biol Trace Elem Res ; 201(5): 2303-2310, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-35852674

RESUMO

Macrophages are important innate immune cells which can be polarized into heterogeneous populations. The inflammatory-activated M1 cells are known to be involved in all kinds of inflammatory diseases, which were also found to be associated with dysregulation of iron metabolism. While iron overload is known to induce M1 polarization, the valence states of iron and its intracellular dynamics during macrophage inflammatory activation have not been identified. In this study, THP-1-derived macrophages were polarized into M1, M2a, M2b, M2c, and M2d cells, and intracellular ferrous iron (Fe(II)) was measured by our previously developed ultrasensitive Fe(II) fluorescent probe. Significant accumulation of Fe(II) was only observed in M1 cells, which was different from the alterations of total iron. Time-dependent change of intracellular Fe(II) during the inflammatory activation was also consistent with the expression shifts of transferrin receptor CD71, ferrireductase Steap3, and Fe(II) exporter Slc40a1. In addition, accumulation of Fe(II) was also found in the colon macrophages of mice with ulcerative colitis, which was positively correlated to inflammatory phenotypes, including the productions of NO, IL-1ß, TNF-α, and IL-6. Collectively, these results demonstrated the specific accumulation of Fe(II) in inflammatory-activated macrophages, which not only enriched our understanding of iron homeostasis in macrophages, but also indicated that Fe(II) could be further developed as a potential biomarker for inflammatory-activated macrophages.


Assuntos
Sobrecarga de Ferro , Ferro , Animais , Camundongos , Ferro/metabolismo , Macrófagos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Sobrecarga de Ferro/metabolismo , Compostos Ferrosos/metabolismo
15.
J Cardiovasc Dev Dis ; 10(5)2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37233166

RESUMO

INTRODUCTION: The COVID-19 pandemic has changed the immunological status of the population, indicating increased activation. The aim of the study was to compare the degree of inflammatory activation in patients admitted for surgical revascularization in the period before and during the COVID-19 pandemic. MATERIALS AND METHODS: This retrospective analysis included an analysis of inflammatory activation assessed on the basis of whole blood counts in 533 patients (435 (82%) male and 98 (18%) female) with a median age of 66 (61-71) years who underwent surgical revascularization, including 343 and 190 patients operated on in 2018 and 2022, respectively. RESULTS: The compared groups were matched by propensity score matching analysis, obtaining 190 patients in each group. Significantly higher values of preoperative monocyte count (p = 0.015), monocyte-to-lymphocyte ratio (p = 0.004) and systemic inflammatory response index (p = 0.022) were found in the during-COVID subgroup. The perioperative and 12-month mortality rates were comparable, with 1% (n = 4) in 2018 vs. 1% (n = 2) in 2022 (p = 0.911), and 5.6 % (n = 11 patients) vs. 7% (n = 13 patients) (p = 0.413), in the pre-COVID and during-COVID subgroups, respectively. CONCLUSIONS: Simple whole blood analysis in patients with complex coronary artery disease performed before and during the COVID-19 pandemic indicates excessive inflammatory activation. However, the immune variation did not interfere with one-year mortality rate after surgical revascularization.

16.
Mult Scler Relat Disord ; 70: 104504, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36623394

RESUMO

BACKGROUND AND OBJECTIVES: Aminoacyl-tRNA synthetase complex interacting with multifunctional protein-1 (AIMP1) has been reported to carry pro-inflammatory properties and anti-angiogenesis effects. However, the exact role of AIMP1 in patients with NMOSD is not yet clear. Our objective was to investigate the relationship between plasma AIMP1 levels and disease severity in patients with AQP4-IgG+ NMOSD from North China based on the Expanded Disability Status Scale (EDSS) score. METHODS: Plasma AIMP1 levels were measured using ELISA kits in 94 patients with AQP4-IgG+NMOSD (48 in the acute phase before high-dose intravenous methylprednisolone (IVMP) therapy, 21 in the acute phase after IVMP therapy, 25 in the clinical remission-phase)as well as 33 healthy controls (HCs). The disability function of NMOSD patients was evaluated using the EDSS score. Furthermore, the clinical characteristics of the patients were also evaluated, and laboratory tests were performed on blood samples. RESULTS: The plasma AIMP1 levels in AQP4-IgG+NMOSD patients with acute phase before IVMP therapy were significantly higher as compared to those in patients after the IVMP therapy (p < 0.001) as well as those in the clinical remission phase (p = 0.021) or HCs (p < 0.001). Plasma AIMP1 levels were positively correlated with EDSS scores (r = 0.485, p < 0.001) and negatively correlated with serum complement 3 concentrations (r =-0.452, p = 0.001). AIMP1 exhibited the potential to distinguish NMOSD from HCs (AUROC 0.820, p < 0.0001) and could differentiate mild and moderate-severe NMOSD (AUROC 0.790, p = 0.0006). Furthermore, plasma AIMP1 levels of ≥49.55pg/mL were found to be an independent predictor of the risk for moderate-severe NMOSD (with OR 0.03, 95%CI 0.001-0.654, p = 0.026). CONCLUSION: AIMP1 may be involved in the pathogenesis of AQP4-IgG+NMOSD disease and predict the disease activity, severity, or effect of treatment in patients with NMOSD. Further studies should be performed to reveal the precise mechanisms of AQP4-IgG+NMOSD.


Assuntos
Neuromielite Óptica , Humanos , Aquaporina 4 , Autoanticorpos , Imunoglobulina G , Metilprednisolona , Neuromielite Óptica/terapia
17.
J Ethnopharmacol ; 309: 116283, 2023 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-36898449

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Jieduquyuziyin prescription (JP), as a traditional Chinese medicine formula, is extensively applied to treat systemic lupus erythematosus (SLE). Its prescription is based on clinical practice and an evidence-based application of traditional medicines. It is approved by use in Chinese hospitals as a clinical prescription that can be directly used. AIM OF THE STUDY: The study aims to elucidate JP's efficacy on lupus-like disease combined with atherosclerosis and to explore its mechanism. MATERIALS AND METHODS: To conduct in vivo experiments, we established a model of lupus-like disease with atherosclerosis in ApoE-/- mice fed a high-fat diet and injected intraperitoneally with pristane. In addition, oxidized low-density lipoprotein (ox-LDL) and a TLR9 agonist (CpG-ODN2395) were utilized to examine the mechanism of JP on SLE combined with AS in RAW264.7 macrophages in vitro. RESULTS: Results indicated that JP reduced hair loss and levels of the spleen index, maintained stable body weight, alleviated kidney damage in mice, and reduced the expression levels of urinary protein, autoantibodies, and inflammatory factors in serum. Furthermore, JP is effective at alleviating the lupus-like symptoms observed in mice. In mice, JP inhibited aortic plaque deposition, stimulated lipid metabolism, and increased the expression of genes that regulate cholesterol efflux, including ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette subfamily G member 1 (ABCG1), scavenger receptor class B type I (SR-BI), and peroxisome proliferator-activated receptor γ (PPAR-γ). In vivo, JP inhibited the expression of the Toll-like receptor 9 (TLR9)-induced signaling pathway, which links TLR9/MyD88/NF-kB to the expression of subsequent inflammatory factors. Furthermore, JP inhibited the expression of TLR9 and MyD88 in vitro. In addition, the JP treatment effectively reduced foam cell formation in RAW264.7 macrophages by increasing the expression of ABCA1/G1, PPAR-γ and SR-BI. CONCLUSIONS: JP played a therapeutic role in ApoE-/- mice with pristane-induced lupus-like diseases and AS, possibly through inhibition of TLR9/MyD88 signaling and promotion of cholesterol efflux.


Assuntos
Aterosclerose , Lúpus Eritematoso Sistêmico , Camundongos , Animais , Receptor Toll-Like 9/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Colesterol/metabolismo , Lipoproteínas LDL/metabolismo , Células Espumosas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aterosclerose/genética , PPAR gama/metabolismo , Apolipoproteínas E/genética , Lúpus Eritematoso Sistêmico/tratamento farmacológico
18.
Basic Clin Neurosci ; 13(1): 117-128, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36589018

RESUMO

Introduction: Introduction: blood-brain-barrier perfusion characterization impaired in MS as some studies have shown recently but a comparison between perfusion parameters in contrast-enhanced and non-enhanced lesions not have been well documented. Pharmacokinetic quantitative parameters have obtained from dynamic contrast-enhanced in magnetic resonance imaging is a useful way to quantify blood-brain barrier permeability leakage. Methods: MR examination was performed on 28 patients with Relapsing-remitted Multiple Sclerosis (RRMS) with (Mean±SD age: 34.7±9.28) which had multiple lesions in the brain.3D dynamic T1-weighted spoiled gradient echo was obtained and Perfusion parameters and its map assessed in enhanced and non-enhanced lesions after intravascular injection differences in parameters and map obtained by analyzing ROI in Extended Toft model. Results: permeability as measured Krtans was a significantly higher value in CE to compare NE lesions. Ktrans and Kep have significant differences in NAWM and CE and NE lesions. Vb was slightly different in NE and CE lesions. Conclusion: Permeability measured as Ktrans was the good parameter to show permeability impairment of BBB in CE lesions. Dysregulation in BBB is an acceptable sign to indicate existence inflammation in CE lesions. Highlights: Multiple Sclerosis,Inflammation,Blood-brain-barrier dysregulation. Plain Language Summary: Inflammation activity in MS patients has an important role to cause BBB dysfunction.in this article to achieve results to confirm the inflammation importance in MS patients with acute lesions. MRI modality have been used and with comparison between acute and chronic lesions and NAWM of MS patient's presence of inflammation have been proved.

19.
Front Mol Neurosci ; 15: 931753, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36172261

RESUMO

Subarachnoid hemorrhage (SAH) is a major cause of death and morbidity worldwide, often due to rupture of intracranial aneurysms (IAs). Immune infiltration and inflammatory activation play key roles in the process of aneurysmal SAH (aSAH). This study aimed to elaborate the immune infiltration and identify related biomarkers both in blood and tissue samples from patients with aSAH. Expression data of aSAH and healthy control samples were obtained from gene expression omnibus (GEO) database. Overall, a blood sample dataset GSE36791 and a tissue sample dataset GSE122897 were included. Differentially expressed genes (DEGs) between aSAH and healthy samples were explored. We applied GO biological and Gene Set Enrichment Analyses (GSEA) processes to access the functional enrichment. Then feature elimination algorithms based on random forest were used to screen and verify the biomarkers of aSAH. We performed three computational algorithms including Cell type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT), Microenvironment Cell Populations-counter (MCPcounter), and xcell to evaluate the immune cell infiltration landscape to identify the unique infiltration characteristics associated with rupturing. We found 2,220 DEGs (856 upregulated and 1,364 downregulated) in the original dataset. Functional analysis revealed most of these genes are enriched in immunological process, especially related with neutrophil response. Similar signaling pathway enrichment patterns were observed in tissue sample dataset and ClueGo. Analysis of immune microenvironment infiltration suggested neutrophils were abnormally upregulated in aSAH compared with those in the control group. Key gene SRPK1 was then filtered based on feature elimination algorithms, and transcription factor (TF) ZNF281 is assumed to participate in immunomodulation by regulating expression of SRPK1. Several immunomodulators such as CXCR1 and CXCR2 also appear to be involved in the progression of aSAH. In the present study, we performed a comprehensive stratification and quantification of the immune infiltration status of aSAH. By exploring the potential mechanism for aSAH based on several computational algorithms, key genes including SRPK1 and ZNF281 were filtered. This study may be of benefit to patients who are at high risk of suffering aSAH which allows for early diagnosis and potential therapy.

20.
Polymers (Basel) ; 14(3)2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35160591

RESUMO

Collagen, as the main component of connective tissue, is frequently used in various tissue engineering applications. In this study, porous sponge-like collagen scaffolds were prepared by freeze-drying and were then mineralized in a simulated body fluid. The mechanical stability was similar in both types of scaffolds, but the mineralized scaffolds (MCS) contained significantly more calcium, magnesium and phosphorus than the unmineralized scaffolds (UCS). Although the MCS contained a lower percentage (~32.5%) of pores suitable for cell ingrowth (113-357 µm in diameter) than the UCS (~70%), the number of human-osteoblast-like MG-63 cells on days 1, 3 and 7 after seeding was higher on MCS than on UCS, and the cells penetrated deeper into the MCS. The cell growth in extracts prepared by eluting the scaffolds for 7 days in a cell culture medium was also markedly higher in the MCS extracts, as indicated by real-time monitoring in the sensory xCELLigence system for 7 days. From this point of view, MCS are more promising for bone tissue engineering than UCS. However, MCS evoked a more pronounced inflammatory response than UCS, as indicated by the production of tumor necrosis factor-alpha (TNF-α) in macrophage-like RAW 264.7 cells in cultures on these scaffolds.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA