Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 697
Filtrar
1.
Inflamm Res ; 71(2): 191-204, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35028708

RESUMO

OBJECTIVE: Inflammation is central to the development and progression of diabetic nephropathy (DN). Although the exact mechanisms of inflammation in the kidney have not been well elucidated, pyrin domain containing 3 (NLRP3) inflammasome activation is involved in the onset and progression of DN. Here, we investigated the underlying regulatory mechanisms of hyperglycaemia-induced NLRP3 inflammasome activation in the kidney. METHODS: HEK293T cells received high glucose, and the cell proliferation and apoptosis were detected. Biochemical indicators in db/db mice were tested by kits, and the morphological changes in the kidney were observed using staining methods and transmission electron microscopy. The interaction of Ras-related C3 botulinum toxin substrate 1 (RAC1) and NLRP3 inflammasome in cells and in mice was assessed by co-immunoprecipitation (Co-IP) and immunofluorescence. Expression of all proteins was examined by western blotting and immunohistochemistry. In additional, the directly combination of RAC1 and NLRP3 was evaluated by GST Pulldown. RESULTS: High-glucose and hyperglycaemia conditions resulted in Ras-related C3 botulinum toxin substrate 1 (RAC1) and NLRP3 inflammasome interactions in cells and in mice. Additionally, RAC1 promoted NLRP3 inflammasome activation and then induced cell damage, and morphological and functional abnormalities in the kidney. We also observed that RAC1 activates the NLRP3 inflammasome by directly binding to NLRP3. CONCLUSION: In the present study, we confirmed that RAC1 binding to NLRP3 is sufficient to activate the NLRP3 inflammasome in the kidney and accelerate DN pathological processes. These results elucidate the upstream cellular and molecular mechanisms of NLRP3 inflammasome activation and provide new therapeutic strategies for the treatment of DN.


Assuntos
Nefropatias Diabéticas/etiologia , Inflamassomos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Animais , Caspase 1/fisiologia , Células HEK293 , Humanos , Hiperglicemia/complicações , Interleucina-1beta/fisiologia , Rim/patologia , Masculino , Camundongos , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
2.
Hum Cell ; 35(1): 226-237, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34591282

RESUMO

Glioblastoma is a glioma characterized by highly malignant features. Numerous studies conducted on the relationship between glioblastoma and the microenvironment have indicated the significance of tumor-associated macrophages/microglia (TAMs) in glioblastoma progression. Since interleukin (IL)-1ß secreted by TAMs has been suggested to promote glioblastoma growth, we attempted to elucidate the detailed mechanisms of IL-1ß in glioblastoma growth in this study. A phospho-receptor tyrosine kinase array and RNA-sequencing studies indicated that IL-1ß induced the activation of signal transducer and activator of transcription-3 and nuclear factor-kappa B signaling. Glioblastoma cells stimulated by IL-1ß induced the production of IL-6 and CXCL8, which synergistically promoted glioblastoma growth via signal transducer and activator of transcription-3 and nuclear factor-kappa B signaling. By immunohistochemistry, IL-1ß expression was seen on TAMs, especially in perinecrotic areas. These results suggest that IL-1ß might be a useful target molecule for anti-glioblastoma therapy.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Glioblastoma/genética , Glioblastoma/patologia , Interleucina-1beta/fisiologia , Macrófagos/metabolismo , Microglia/metabolismo , NF-kappa B/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Neoplasias Encefálicas/terapia , Linhagem Celular , Expressão Gênica , Glioblastoma/terapia , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Terapia de Alvo Molecular
3.
Sci Rep ; 11(1): 21697, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34737366

RESUMO

IKKα and IKKß are essential kinases for activating NF-κB transcription factors that regulate cellular differentiation and inflammation. By virtue of their small size, chemokines support the crosstalk between cartilage and other joint compartments and contribute to immune cell chemotaxis in osteoarthritis (OA). Here we employed shRNA retroviruses to stably and efficiently ablate the expression of each IKK in primary OA chondrocytes to determine their individual contributions for monocyte chemotaxis in response to chondrocyte conditioned media. Both IKKα and IKKß KDs blunted both the monocyte chemotactic potential and the protein levels of CCL2/MCP-1, the chemokine with the highest concentration and the strongest association with monocyte chemotaxis. These findings were mirrored by gene expression analysis indicating that the lowest levels of CCL2/MCP-1 and other monocyte-active chemokines were in IKKαKD cells under both basal and IL-1ß stimulated conditions. We find that in their response to IL-1ß stimulation IKKαKD primary OA chondrocytes have reduced levels of phosphorylated NFkappaB p65pSer536 and H3pSer10. Confocal microscopy analysis revealed co-localized p65 and H3pSer10 nuclear signals in agreement with our findings that IKKαKD effectively blunts their basal level and IL-1ß dependent increases. Our results suggest that IKKα could be a novel OA disease target.


Assuntos
Quinase I-kappa B/metabolismo , Interleucina-1beta/metabolismo , Monócitos/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocinas/imunologia , Quimiocinas/metabolismo , Quimiotaxia/fisiologia , Condrócitos/metabolismo , Feminino , Humanos , Quinase I-kappa B/fisiologia , Inflamação , Interleucina-1beta/fisiologia , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Osteoartrite/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases , Transdução de Sinais/fisiologia , Fator de Transcrição RelA
4.
Int Immunopharmacol ; 101(Pt A): 108323, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34749292

RESUMO

BACKGROUND: End stage renal disease (ESRD) has caused public health problem with high prevalence worldwide. Peritoneum from peritoneal dialysis patients with ESRD can induce pathological changes of the peritoneum, including fibrosis. The trans-differentiation of pericytes has been found to be closely associated with inflammatory diseases, such as organ fibrosis. However, the function of macrophages in regulating the transition of pericyte to peritoneal fibrosis is unclear. METHODS: Histological examination was conducted using Hematoxylin and eosin (HE) staining and Masson's trichrome staining. The protein levels were determined via western blot. Enzyme-linked immunosorbent assay (ELISA) was used to examine IL-1ß concentrations. Gasdermin D (GSDMD) was knocked out in mice by Clustered Regularly Interspaced Short Palindromic Repeats-CRISPR-Associated 9 (CRISPR-Cas9). RESULTS: Mice receiving dextrose peritoneal dialysate displayed mesothelial cell monolayer loss and thickness of submesothelial compact zone increase. Moreover, dextrose peritoneal dialysate treatment up-regulated GSDMD expression. GSDMD knockdown inhibited IL-1ß production in macrophages. Further, pericytes were treated with cultural supernatant from macrophages. We found that GSDMD knockdown suppressed fibrosis and vascular endothelial growth factor (VEGF)/phosphoinositide 3-kinase (PI3K) pathway in pericytes. In addition, GSDMD were knocked out in mice using CRISPR/Cas9. The histological examinations revealed that GSDMD-/- alleviated the damage of peritoneal tissue and thickness of submesothelial compact zone. GSDMD-/- attenuated interleukin-1beta (IL-1ß) level and peritoneal fibrosis induced by dextrose peritoneal dialysate treatment in pericytes in vivo. CONCLUSION: These results demonstrated that macrophages can regulate the transition of pericyte to peritoneal fibrosis via the GSDMD/IL-1ß axis, which provides a new therapeutic target.


Assuntos
Transição Epitelial-Mesenquimal , Interleucina-1beta/metabolismo , Macrófagos/fisiologia , Pericitos/fisiologia , Fibrose Peritoneal/etiologia , Proteínas de Ligação a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Animais , Western Blotting , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Citometria de Fluxo , Imunofluorescência , Técnicas de Silenciamento de Genes , Interleucina-1beta/fisiologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Pericitos/metabolismo , Fibrose Peritoneal/metabolismo , Proteínas de Ligação a Fosfato/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia
5.
Cells ; 10(9)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34571989

RESUMO

Hypoxia and inflammation are frequently co-incidental features of the tissue microenvironment in a wide range of inflammatory diseases. While the impact of hypoxia on inflammatory pathways in immune cells has been well characterized, less is known about how inflammatory stimuli such as cytokines impact upon the canonical hypoxia-inducible factor (HIF) pathway, the master regulator of the cellular response to hypoxia. In this review, we discuss what is known about the impact of two major pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), on the regulation of HIF-dependent signaling at sites of inflammation. We report extensive evidence for these cytokines directly impacting upon HIF signaling through the regulation of HIF at transcriptional and post-translational levels. We conclude that multi-level crosstalk between inflammatory and hypoxic signaling pathways plays an important role in shaping the nature and degree of inflammation occurring at hypoxic sites.


Assuntos
Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-1beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Citocinas/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Hipóxia/fisiopatologia , Fator 1 Induzível por Hipóxia/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Inflamação/fisiopatologia , Interleucina-1beta/fisiologia , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Ativação Transcricional , Fator de Necrose Tumoral alfa/fisiologia
6.
Biol Psychol ; 165: 108147, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34492333

RESUMO

Although elevations in systemic suPAR levels have been associated with inflammatory conditions and with exposure to life stress and adversity, it is not yet clear whether acute psychological stress influences suPAR levels, either systemically and/or in saliva. The aim of this study was to investigate whether salivary suPAR levels are increased following exposure to acute psychological stress. Healthy subjects, aged 18-40 years, completed a laboratory psychological stressor and provided saliva samples before and after the stress test (60 min apart). Levels of suPAR as well as those of cytokines increased in the post-stress samples (all ps < .001). Baseline and post-stress IL-1ß and TNF-α as well as post-stress IL-6 correlated significantly with suPAR (all ps < .01), but IL-10 and baseline IL-6 did not. These results show that suPAR levels in saliva are stress-reactive and suggest a potential application as stress biomarkers in saliva, particularly given the advantage of easily detectable concentrations.


Assuntos
Receptores de Ativador de Plasminogênio Tipo Uroquinase , Saliva , Estresse Psicológico , Biomarcadores , Humanos , Interleucina-10/sangue , Interleucina-10/fisiologia , Interleucina-1beta/sangue , Interleucina-1beta/fisiologia , Interleucina-6/sangue , Interleucina-6/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Fator de Necrose Tumoral alfa/sangue
7.
J Biol Chem ; 297(3): 100987, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34364871

RESUMO

Progressive fibrosis leads to loss of organ function and affects many organs as a result of excessive extracellular matrix production. The ubiquitous matrix polysaccharide hyaluronan (HA) is central to this through association with its primary receptor, CD44, which exists as standard CD44 (CD44s) or multiple splice variants. Mediators such as profibrotic transforming growth factor (TGF)-ß1 and proinflammatory interleukin (IL)-1ß are widely associated with fibrotic progression. TGF-ß1 induces myofibroblast differentiation, while IL-1ß induces a proinflammatory fibroblast phenotype that promotes fibroblast binding to monocyte/macrophages. CD44 expression is essential for both responses. Potential CD44 splice variants involved, however, are unidentified. The TGF-ß1-activated CD44/epidermal growth factor receptor complex induces differentiation of metastatic cells through interactions with the matrix metalloproteinase inducer, CD147. This study aimed to determine the CD44 variants involved in TGF-ß1- and IL-1ß-mediated responses and to investigate the potential profibrotic role of CD147. Using immunocytochemistry and quantitative PCR, standard CD44s were shown to be essential for both TGF-ß1-induced fibroblast/myofibroblast differentiation and IL-1ß-induced monocyte binding. Co-immunoprecipitation identified that CD147 associated with CD44s. Using CD147-siRNA and confocal microscopy, we also determined that incorporation of the myofibroblast marker, αSMA, into F-actin stress fibers was prevented in the absence of CD147 and myofibroblast-dependent collagen gel contraction was inhibited. CD147 did not associate with HA, but removal of HA prevented the association of CD44s with CD147 at points of cell-cell contact. Taken together, our data suggest that CD44s/CD147 colocalization is essential in regulating the mechanical tension required for the αSMA incorporation into F-actin stress fibers that regulates myofibroblast phenotype.


Assuntos
Basigina/fisiologia , Diferenciação Celular/fisiologia , Receptores de Hialuronatos/fisiologia , Miofibroblastos/citologia , Fator de Crescimento Transformador beta1/fisiologia , Basigina/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Interleucina-1beta/fisiologia , Miofibroblastos/metabolismo
8.
Int J Mol Sci ; 22(16)2021 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-34445357

RESUMO

The biological activities of interleukins, a group of circulating cytokines, are linked to the immuno-pathways involved in many diseases. Mounting evidence suggests that interleukin-1ß (IL-1ß) plays a significant role in the pathogenesis of various types of hypertension. In this review, we summarized recent findings linking IL-1ß to systemic arterial hypertension, pulmonary hypertension, and gestational hypertension. We also outlined the new progress in elucidating the potential mechanisms of IL-1ß in hypertension, focusing on it's regulation in inflammation, vascular smooth muscle cell function, and extracellular remodeling. In addition, we reviewed recent studies that highlight novel findings examining the function of non-coding RNAs in regulating the activity of IL-1ß and its associated proteins in the setting of hypertension. The information collected in this review provides new insights into understanding the pathogenesis of hypertension and could lead to the discovery of new anti-hypertensive therapies to combat this highly prevalent disease.


Assuntos
Hipertensão/etiologia , Interleucina-1beta/fisiologia , Animais , Matriz Extracelular/patologia , Matriz Extracelular/fisiologia , Feminino , Regulação da Expressão Gênica , Humanos , Hipertensão/patologia , Hipertensão/fisiopatologia , Hipertensão Induzida pela Gravidez/etiologia , Hipertensão Induzida pela Gravidez/patologia , Hipertensão Induzida pela Gravidez/fisiopatologia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Inflamação/complicações , Inflamação/fisiopatologia , Interleucina-1beta/genética , Músculo Liso Vascular/fisiopatologia , Gravidez , RNA não Traduzido/fisiologia , Remodelação Vascular/fisiologia
9.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-34201546

RESUMO

Tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) are two cytokines involved in the perpetuation of the chronic inflammation state characterizing rheumatoid arthritis (RA). Significant advances in the treatment of this pathology have been made over the past ten years, partially through the development of anti-TNF and anti-IL-1 therapies. However, major side effects still persist and new alternative therapies should be considered. The formulation of the micro-immunotherapy medicine (MIM) 2LARTH® uses ultra-low doses (ULD) of TNF-α, IL-1ß, and IL-2, in association with other immune factors, to gently restore the body's homeostasis. The first part of this review aims at delineating the pivotal roles played by IL-1ß and TNF-α in RA physiopathology, leading to the development of anti-TNF and anti-IL-1 therapeutic agents. In a second part, an emphasis will be made on explaining the rationale of using multiple therapeutic targets, including both IL-1ß and TNF-α in 2LARTH® medicine. Particular attention will be paid to the ULD of those two main pro-inflammatory factors in order to counteract their overexpression through the lens of their molecular implication in RA pathogenesis.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Citocinas/administração & dosagem , Imunoterapia/métodos , Interleucina-1beta/administração & dosagem , Fator de Necrose Tumoral alfa/administração & dosagem , Administração Oral , Animais , Artrite Reumatoide/fisiopatologia , Relação Dose-Resposta a Droga , Humanos , Interleucina-1beta/efeitos adversos , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/fisiologia , Interleucina-2/administração & dosagem , Interleucina-2/efeitos adversos , Terapia de Alvo Molecular/métodos , Medicina de Precisão , Fator de Necrose Tumoral alfa/efeitos adversos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia
10.
Front Immunol ; 12: 643149, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34177893

RESUMO

Fibrosis is the final common pathway of inflammatory diseases in various organs. The inflammasomes play an important role in the progression of fibrosis as innate immune receptors. There are four main members of the inflammasomes, such as NOD-like receptor protein 1 (NLRP1), NOD-like receptor protein 3 (NLRP3), NOD-like receptor C4 (NLRC4), and absent in melanoma 2 (AIM2), among which NLRP3 inflammasome is the most studied. NLRP3 inflammasome is typically composed of NLRP3, ASC and pro-caspase-1. The activation of inflammasome involves both "classical" and "non-classical" pathways and the former pathway is better understood. The "classical" activation pathway of inflammasome is that the backbone protein is activated by endogenous/exogenous stimulation, leading to inflammasome assembly. After the formation of "classic" inflammasome, pro-caspase-1 could self-activate. Caspase-1 cleaves cytokine precursors into mature cytokines, which are secreted extracellularly. At present, the "non-classical" activation pathway of inflammasome has not formed a unified model for activation process. This article reviews the role of NLRP1, NLRP3, NLRC4, AIM2 inflammasome, Caspase-1, IL-1ß, IL-18 and IL-33 in the fibrogenesis.


Assuntos
Fibrose/etiologia , Inflamassomos/fisiologia , Animais , Proteínas Adaptadoras de Sinalização CARD/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Caspase 1/fisiologia , Humanos , Inflamassomos/classificação , Interleucina-1beta/fisiologia , Interleucina-33/fisiologia , Rim/patologia , Cirrose Hepática/etiologia , Miocárdio/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas NLR/fisiologia , Fibrose Pulmonar/etiologia
11.
Cell Host Microbe ; 29(6): 959-974.e7, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-33894128

RESUMO

Microbiota play critical roles in regulating colitis and colorectal cancer (CRC). However, it is unclear how the microbiota generate protective immunity against these disease states. Here, we find that loss of the innate and adaptive immune signaling molecule, TAK1, in myeloid cells (Tak1ΔM/ΔM) yields complete resistance to chemical-induced colitis and CRC through microbiome alterations that drive protective immunity. Tak1ΔM/ΔM mice exhibit altered microbiota that are critical for resistance, with antibiotic-mediated disruption ablating protection and Tak1ΔM/ΔM microbiota transfer conferring protection against colitis or CRC. The altered microbiota of Tak1ΔM/ΔM mice promote IL-1ß and IL-6 signaling pathways, which are required for induction of protective intestinal Th17 cells and resistance. Specifically, Odoribacter splanchnicus is abundant in Tak1ΔM/ΔM mice and sufficient to induce intestinal Th17 cell development and confer resistance against colitis and CRC in wild-type mice. These findings identify specific microbiota strains and immune mechanisms that protect against colitis and CRC.


Assuntos
Bacteroidetes/metabolismo , Colite/microbiologia , Neoplasias Colorretais/microbiologia , Citocinas/fisiologia , Microbioma Gastrointestinal , MAP Quinase Quinase Quinases/fisiologia , Células Th17/metabolismo , Animais , Colite/induzido quimicamente , Colite/metabolismo , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/metabolismo , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Interações entre Hospedeiro e Microrganismos , Imunidade Inata , Interleucina-1beta/fisiologia , Interleucina-6/fisiologia , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Transdução de Sinais , Células Th17/imunologia
12.
Br J Cancer ; 124(11): 1754-1756, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33758330

RESUMO

Immune checkpoint blockade (ICB) has demonstrated efficacy in multiple cancers, offering the potential of long-term disease control not achievable with cytotoxic or targeted therapies. However, the field has not yet achieved the crucial next steps - the expansion of the response rate and achievement of clinical efficacy in so-called "cold tumours". Mechanistic studies of tumour-type specific immunosuppressive pathways can reveal underlying biological hurdles to immunotherapy and offer new therapeutic insights. Our finding that tumour-derived IL-1ß mediates immunosuppression in pancreatic cancer has precipitated a new clinical trial.


Assuntos
Tolerância Imunológica/genética , Imunoterapia , Interleucina-1beta/fisiologia , Neoplasias Pancreáticas/terapia , Antineoplásicos Imunológicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Humanos , Inibidores de Checkpoint Imunológico/administração & dosagem , Imunoterapia/métodos , Imunoterapia/tendências , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
13.
Int Immunopharmacol ; 93: 107388, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33529913

RESUMO

Endothelial dysfunction is a typical characteristic of sepsis. Endothelial nitric oxide synthase (eNOS) is important for maintaining endothelial function. Our previous study reported that the NLRP3 inflammasome promoted endothelial dysfunction by enhancing inflammation. However, the effects of NLRP3 on eNOS require further investigation. Therefore, the present study aimed to investigate the role of NLRP3 on eNOS expression levels in cecal ligation and puncture-induced impaired endothelium-dependent vascular relaxation and to determine the protective effects of melatonin. eNOS expression levels were discovered to be downregulated in the mesenteric arteries of sepsis model mice. Inhibiting NLRP3 with 10 mg/ kg MCC950 or inhibiting IL-1ß with 100 mg diacerein rescued the eNOS expression and improved endothelium-dependent vascular relaxation. In vitro, IL-1ß stimulation downregulated eNOS expression levels in human aortic endothelial cells (HAECs) in a concentration- and time-dependent manner, while pretreatment with 1 µM of the proteasome inhibitor MG132 reversed this effect. In addition, treatment with 10 mg/kg MG132 also prevented the proteolysis of eNOS and improved endothelium-dependent vascular relaxation in vivo. Notably, treatment with 30 mg/kg melatonin downregulated NLRP3 expression levels and decreased IL-1ß secretion, subsequently increasing the expression of eNOS and improving endothelium-dependent vascular relaxation. In conclusion, the findings of the present study indicated that the NLRP3/IL-1ß axis may impair vasodilation by promoting the proteolysis of eNOS and melatonin may protect against sepsis-induced endothelial relaxation dysfunction by inhibiting the NLRP3/IL-1ß axis, suggesting its pharmacological potential in sepsis.


Assuntos
Interleucina-1beta/fisiologia , Melatonina/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Sepse/tratamento farmacológico , Vasodilatação/fisiologia , Animais , Aorta/citologia , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Humanos , Masculino , Melatonina/farmacologia , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , Camundongos Endogâmicos C57BL , Proteólise , Sepse/metabolismo , Sepse/fisiopatologia , Vasodilatação/efeitos dos fármacos
14.
J Orthop Surg Res ; 16(1): 40, 2021 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-33430857

RESUMO

BACKGROUND: Knee osteoarthritis (KOA) seriously affects the quality of life of KOA patients. This study aimed to investigate whether miR-107 could regulate KOA through pyroptosis to affect collagen protein secreted by chondrocytes through IL-1ß. METHODS: The proliferation of chondrocytes was detected by CCK-8 assay. RT-qPCR analysis was used to identify miR-107 expression and transfection effects. The expression of Col II, IL-1ß, IL-18, and MMP13 in supernatant of chondrocytes or chondrocytes was detected by ELISA assay and western blot analysis. The pyroptosis of chondrocytes was analyzed by TUNEL assay and the expression of pyroptosis-related proteins was analyzed by western blot. Luciferase reporter assay confirmed the relation of miR-107 to caspase-1. RESULTS: The proliferation of chondrocytes was decreased after LPS induction and further decreased by treatment of ATP. Single LPS treatment for chondrocytes downregulated the Col II expression while upregulated the expression of IL-1ß, IL-18, and MMP-13, which was further changed by ATP treatment. miR-107 expression was decreased in chondrocytes induced by LPS and further decreased in chondrocytes induced by LPS and ATP. In addition, miR-107 overexpression increased the proliferation and decreased the pyroptosis of chondrocytes induced by LPS and ATP. miR-107 overexpression upregulated the Col II expression while down-regulated the expression of IL-1ß, IL-18, and MMP-13 in supernatant of chondrocytes or chondrocytes induced by LPS and ATP. miR-107 overexpression down-regulated the expression of caspase-1, c-caspase-1, GSDMD-N, and TLR4 in chondrocytes induced by LPS and ATP. Furthermore, miR-107 directly targeted caspase-1. CONCLUSIONS: miR-107 can protect against KOA by downregulating caspase-1 to decrease pyroptosis, thereby promoting collagen protein secreted by chondrocytes by down-regulating IL-1ß.


Assuntos
Caspase 1/genética , Caspase 1/metabolismo , Condrócitos/metabolismo , Condrócitos/fisiologia , Regulação da Expressão Gênica/genética , Proteínas Matrilinas/metabolismo , MicroRNAs/fisiologia , Osteoartrite do Joelho/genética , Osteoartrite do Joelho/metabolismo , Proteólise , Proliferação de Células/genética , Células Cultivadas , Colágeno/metabolismo , Regulação para Baixo/genética , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/fisiologia , Osteoartrite do Joelho/patologia , Piroptose/genética
15.
J Mol Med (Berl) ; 99(3): 403-414, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33409553

RESUMO

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with poor prognosis and overall survival. Clinical investigations show that chronic stress is commonly present in the course of AML and associated with adverse outcome. However, the underlying molecular mechanisms are elusive. In the present study, a chronic restraint stress mouse model was established to evaluate the effect of stress on AML. We found that mice under chronic stress exhibited significantly increased liver and spleen infiltration of leukemic cells and poorer overall survival. This was accompanied by elevated cellular NLR family pyrin domain containing 3 (NLRP3) and interleukin-1ß (IL-1ß) in the liver or bone marrow, and secreted IL-1ß in the plasma, indicating the activation of inflammasomes under chronic restraint stress. High mobility group box 1 (HMGB1) expression was markedly increased in newly diagnosed AML patients, but reduced in complete remission AML patients. The expression level of HMGB1 was positively correlated with NLRP3 mRNA in AML patients. Knockdown of HMGB1 significantly decreased NLRP3 and IL-1ß expression in AML cell lines, and secreted IL-1ß in supernatant of AML cell culture, while HMGB1 stimulation caused contrary effects. These results implied that HMGB1 could be involved in the regulation of inflammasome activation in AML development. Mice model showed that chronic restraint stress-facilitated proliferation and infiltration of AML cells were largely abrogated by knocking down HMGB1. Knockdown of HMGB1 also ameliorated overall survival and remarkably neutralized NLRP3 and IL-1ß expression under chronic restraint stress. These findings provide evidences that chronic stress promotes AML progression via HMGB1/NLRP3/IL-1ß dependent mechanism, suggesting that HMGB1 is a potential therapeutic target for AML. KEY MESSAGES: • Chronic restraint stress promoted acute myeloid leukemia (AML) progression and mediated NLRP3 inflammasome activation in xenograft mice. • HMGB1 mediated NLRP3 inflammasome activation in AML cells. • Knockdown of HMGB1 inhibited AML progression under chronic stress in vivo.


Assuntos
Regulação Leucêmica da Expressão Gênica , Proteína HMGB1/fisiologia , Interleucina-1beta/fisiologia , Leucemia Mieloide Aguda/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas de Neoplasias/fisiologia , Transdução de Sinais/fisiologia , Animais , Medula Óssea/metabolismo , Linhagem Celular Tumoral , Doença Crônica , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/biossíntese , Proteína HMGB1/genética , Xenoenxertos , Humanos , Inflamassomos/metabolismo , Inflamação , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Leucemia Mieloide Aguda/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Interferência de RNA , Indução de Remissão , Restrição Física , Baço/metabolismo , Baço/patologia , Estresse Fisiológico , Receptor 4 Toll-Like/fisiologia
16.
J Gastroenterol Hepatol ; 36(3): 740-750, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32918847

RESUMO

BACKGROUND AND AIM: The inflammasomes promote pro-caspase-1 cleavage, leading to processing of pro-interleukin (IL)-1ß into its mature form. We investigated the role of the IL-1ß and nucleotide binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome in gastric injury in mice receiving water-immersion restraint stress (WIRS), focusing on the cyclooxygenase (COX)-2/prostaglandin (PG) E2 axis. METHODS: To induce gastric injury, the mice were placed in a restraint cage and immersed in the water bath to the level of the xiphoid process. Protein levels of mature caspase-1 and IL-1ß were assessed by western blotting. RESULTS: Water-immersion restraint stress induced gastric injury with increase in IL-1ß expression by activation of NLRP3 inflammasome. Exogenous IL-1ß attenuated the injury, whereas anti-IL-1ß neutralizing antibody and IL-1ß receptor antibody aggravated it. NLRP3-/- and caspase-1-/- mice enhanced the injury with reducing of mature IL-1ß, and this aggravation was reduced by exogenous IL-1ß supplementation. Toll-like receptor 4-/- mice were hyporesponsive to WIRS in terms of mature IL-1ß production. Rabeprazole attenuated the injury with preventing inflammasome activation. WIRS injured the stomach with promotion of COX-2 mRNA and PGE2 production, and exogenous IL-1ß enhanced these molecules, while IL-1ß immunoneutralization exerted opposite effect. PGE2 supplementation abolished the hypersensitivity in NLRP3-/- and caspase-1-/- mice through negative regulation of inflammatory cytokines. CONCLUSION: These results suggest that NLRP3 inflammasome-derived IL-1ß plays a protective role in stress-induced gastric injury via activation of the COX-2/PGE2 axis. Toll-like receptor 4 signaling and gastric acid may be involved in NLRP3 inflammasome activation.


Assuntos
Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Gastropatias/etiologia , Gastropatias/prevenção & controle , Estresse Psicológico/complicações , Animais , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ácido Gástrico/metabolismo , Camundongos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Gastropatias/genética , Gastropatias/metabolismo , Receptor 4 Toll-Like/metabolismo
17.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33182538

RESUMO

Pro-inflammatory cytokines like interleukin-1ß (IL-1ß) are upregulated during early responses to tissue damage and are expected to transiently compromise the mechanical microenvironment. Fibroblasts are key regulators of tissue mechanics in the lungs and other organs. However, the effects of IL-1ß on fibroblast mechanics and functions remain unclear. Here we treated human pulmonary fibroblasts from control donors with IL-1ß and used Atomic Force Microscopy to unveil that IL-1ß significantly reduces the stiffness of fibroblasts concomitantly with a downregulation of filamentous actin (F-actin) and alpha-smooth muscle (α-SMA). Likewise, COL1A1 mRNA was reduced, whereas that of collagenases MMP1 and MMP2 were upregulated, favoring a reduction of type-I collagen. These mechanobiology changes were functionally associated with reduced proliferation and enhanced migration upon IL-1ß stimulation, which could facilitate lung repair by drawing fibroblasts to sites of tissue damage. Our observations reveal that IL-1ß may reduce local tissue rigidity by acting both intracellularly and extracellularly through the downregulation of fibroblast contractility and type I collagen deposition, respectively. These IL-1ß-dependent mechanical effects may enhance lung repair further by locally increasing pulmonary tissue compliance to preserve normal lung distension and function. Moreover, our results support that IL-1ß provides innate anti-fibrotic protection that may be relevant during the early stages of lung repair.


Assuntos
Interleucina-1beta/fisiologia , Pulmão/fisiologia , Actinas/metabolismo , Adolescente , Adulto , Fenômenos Biomecânicos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Ciclo-Oxigenase 2/metabolismo , Elasticidade/efeitos dos fármacos , Elasticidade/fisiologia , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Humanos , Interleucina-1beta/farmacologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Masculino , Microscopia de Força Atômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regeneração/genética , Regeneração/fisiologia , Cicatrização/efeitos dos fármacos , Cicatrização/genética , Cicatrização/fisiologia , Adulto Jovem
18.
Hepatology ; 72(4): 1310-1326, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33090557

RESUMO

BACKGROUND AND AIMS: T cells from patients with primary sclerosing cholangitis (PSC) show a prominent interleukin (IL)-17 response upon stimulation with bacteria or fungi, yet the reasons for this dominant T-helper 17 (Th17) response in PSC are not clear. Here, we analyzed the potential role of monocytes in microbial recognition and in skewing the T-cell response toward Th17. APPROACH AND RESULTS: Monocytes and T cells from blood and livers of PSC patients and controls were analyzed ex vivo and in vitro using transwell experiments with cholangiocytes. Cytokine production was measured using flow cytometry, enzyme-linked immunosorbent assay, RNA in situ hybridization, and quantitative real-time PCR. Genetic polymorphisms were obtained from ImmunoChip analysis. Following ex vivo stimulation with phorbol myristate acetate/ionomycin, PSC patients showed significantly increased numbers of IL-17A-producing peripheral blood CD4+ T cells compared to PBC patients and healthy controls, indicating increased Th17 differentiation in vivo. Upon stimulation with microbes, monocytes from PSC patients produced significantly more IL-1ß and IL-6, cytokines known to drive Th17 cell differentiation. Moreover, microbe-activated monocytes induced the secretion of Th17 and monocyte-recruiting chemokines chemokine (C-C motif) ligand (CCL)-20 and CCL-2 in human primary cholangiocytes. In livers of patients with PSC cirrhosis, CD14hiCD16int and CD14loCD16hi monocytes/macrophages were increased compared to alcoholic cirrhosis, and monocytes were found to be located around bile ducts. CONCLUSIONS: PSC patients show increased Th17 differentiation already in vivo. Microbe-stimulated monocytes drive Th17 differentiation in vitro and induce cholangiocytes to produce chemokines mediating recruitment of Th17 cells and more monocytes into portal tracts. Taken together, these results point to a pathogenic role of monocytes in patients with PSC.


Assuntos
Colangite Esclerosante/imunologia , Monócitos/fisiologia , Células Th17/citologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas Adaptadoras de Sinalização CARD/genética , Diferenciação Celular , Quimiocinas/biossíntese , Feminino , Humanos , Interleucina-1beta/fisiologia , Interleucinas/genética , Cirrose Hepática/imunologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
19.
Cell Death Dis ; 11(10): 901, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-33093455

RESUMO

Activation of the nucleotide-binding domain leucine-rich repeat and pyrin domain containing receptor 3 (NLRP3) inflammasome plays an important role in ocular neovascularization. In our study, we found that the expression and activation levels of NLRP3 inflammasome components, including NLRP3, an apoptosis-associated speck-like protein (ASC) containing caspase activation and recruitment domain (CARD) and caspase-1 (CAS1), were significantly upregulated. In addition, we found interleukin (IL)-1ß activity increased while IL-18 activity decreased in the retinas of oxygen-induced ischemic retinopathy (OIR) mice. MCC950, an inhibitor of NLRP3, reversed the IL-1ß/IL-18 activation pattern, inhibited the formation of retinal neovascularization (RNV), decreased the number of acellular capillaries and reduced leakage of retinal vessels. Moreover, MCC950 could regulate the expression of endothelial cell- and pericyte function-associated molecules, such as vascular endothelial growth factor (VEGF), VEGF receptor (VEGFR)1, VEGFR2, matrix metalloproteinase (MMP)2, MMP9, tissue inhibitor of metalloproteinases (TIMP)1, TIMP2, platelet-derived growth factor receptor-ß (PDGFR-ß), platelet-derived growth factor-B (PDGF-B), and angiopoietin2 (Ang2). In vitro, recombinant human (r)IL-18 and rIL-1ß regulated the expression of endothelial cell- and pericyte function-associated molecules and the proliferation and migration of endothelial cells and pericytes. We therefore determined that inhibiting the NLRP3 inflammasome with MCC950 can regulate the function of endothelial cells and pericytes by reversing the IL-1ß/IL-18 activation pattern to ameliorate RNV and leakage; thereby opening new avenues to treat RNV-associated ocular diseases.


Assuntos
Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Inflamassomos/fisiologia , Interleucina-18/fisiologia , Interleucina-1beta/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Sulfonas/farmacologia , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Furanos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Indenos , Isquemia/induzido quimicamente , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Recombinantes , Doenças Retinianas/induzido quimicamente , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Sulfonamidas , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Biochem Biophys Res Commun ; 533(4): 1283-1289, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33066959

RESUMO

We have previously shown a fraction of stromal fibroblasts/myofibroblasts (Fibs) from leukemic bone marrow cells expresses leukemia-specific transcripts along with hematopoietic and Fib-related markers. Normal bone marrow-derived Fibs (nFibs) do not express CD34 or CD45; however, nFibs may express hematopoietic markers with some specific stimulations. CD34 expression was detected in nFib cultures following the addition of a culture supernatant of blood mononuclear cells stimulated with phytohemagglutinin (PHA)-P. To identify the molecules responsible for inducing CD34 expression in nFibs, cDNA clones were isolated using functional expression cloning with a library constructed from PHA-P-stimulated human blood mononuclear cells. Positive clones inducing CD34 transcription in nFibs were selected. We confirmed that an isolated positive cDNA clone encoded human interleukin (IL)-1 beta (ß). CD34 expression was observed in the nFib cultures with recombinant human (rh) IL-1ß protein. And CD34 transcription was suppressed when a rhIL-1ß neutralizing antibody was added to the IL-1ß-stimulated nFib cultures. nFibs expressed gp130 and IL-6 receptors, and CD45 expression was detected in nFibs cultured with rhIL-1ß and rhIL-6. Chronic myelogenous leukemia (CML) cells reportedly respond well to IL-1ß. When CML-derived Fibs were cultured with rhIL-1ß and rhIL-6, CD45-positive cells increased in number. Cell fate may be influenced by an external specific stimulation without gene introduction.


Assuntos
Antígenos CD34/biossíntese , Células da Medula Óssea/metabolismo , Miofibroblastos/metabolismo , Células da Medula Óssea/citologia , Clonagem Molecular , Citocinas/biossíntese , Humanos , Interleucina-1beta/fisiologia , Interleucina-6/fisiologia , Antígenos Comuns de Leucócito/biossíntese , Miofibroblastos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...