Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biomaterials ; 305: 122466, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38184960

RESUMO

Inflammation is associated with a series of diseases like cancer, cardiovascular disease and infection, and phosphorylation/dephosphorylation modification of proteins are important in inflammation regulation. Here we designed and synthesized a novel Brazilin-Ce nanoparticle (BX-Ce NPs) using Brazilin, which has been used for anti-inflammation in cardiovascular diseases but with narrow therapeutic window, and Cerium (IV), a lanthanide which has the general activity in catalyzing the hydrolysis of phosphoester bonds, to conferring de/anti-phosphorylation of IKKß. We found that BX-Ce NPs specifically bound to Asn225 and Lys428 of IKKß and inhibited its phosphorylation at Ser181, contributing to appreciably anti-inflammatory effect in cellulo (IC50 = 2.5 µM). In vivo mouse models of myocardial infarction and sepsis also showed that the BX-Ce NPs significantly ameliorated myocardial injury and improved survival in mice with experimental sepsis through downregulating phosphorylation of IKKß. These findings provided insights for developing metal nanoparticles for guided ion interfere therapy, particularly synergistically target de/anti-phosphorylation as promising therapeutic agents for inflammation and related diseases.


Assuntos
Benzopiranos , Cério , Nanopartículas Metálicas , Nanopartículas , Sepse , Camundongos , Animais , Fosforilação , Quinase I-kappa B/metabolismo , Quinase I-kappa B/uso terapêutico , Inflamação/tratamento farmacológico , Nanopartículas/química , Nanopartículas Metálicas/uso terapêutico , Cério/química
2.
Integr Cancer Ther ; 22: 15347354231172732, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37157810

RESUMO

6-Shogaol from ginger has anti-inflammatory, anti-oxidation and anti-cancer effects. Aim of the Study: To study the effects and possible mechanisms of 6-Shogaol on inhibiting the migration of colon cancer cells Caco2 and HCT116 and prove the effects on proliferation and apoptosis. Materials and methods: The cells were treated with 6-Shogaol at the concentrations of 20, 40, 60, 80, and 100 µM, the cytotoxicity was tested by Colony formation assays and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and the Western blot was used to evaluate IKKß/NF-κB/Snail pathway and EMT-related proteins. In addition, in order to eliminate the interference of proliferation inhibition on the experiment, Caco2 cells were treated with 6-Shogaol at the concentrations of 0, 40, and 80 µM, HCT116 cells were treated with 6-Shogaol at the concentrations of 0, 20, and 40 µM, apoptosis was measured by Annex V/PI staining, and migration was measured by Wound healing assays and Transwell test. Results: 6-Shogaol significantly inhibited the growth of cells. The maximum inhibitory concentration of half of them was 86.63 µM in Caco2 cells and 45.25 µM in HCT116 cells. At 80 µM and 40 µM concentrations, 6-Shogaol significantly promoted apoptosis of colon cancer Caco2 cells and HCT116 cells, and also significantly inhibited cell migration (P < .05). In addition, Western blot analysis showed that at 80 µM dose of 6-Shogaol significantly reduced MMP-2, N-cadherin, IKKß, P-NF-κB and Snail expression in Caco2 cells (P < .05). 40 µM dose of 6-Shogaol significantly reduced VEGF, IKKß, and P-NF-κB expression, and MMP-2, N-cadherin and Snail was significantly decreased at 60 µM of 6-Shogaol in HCT116 cells(P < .05). However, there was no significant change in E-cadherin in Caco2 cells, and the expression of E-cadherin protein in HCT116 cells decreased. Conclusion: This study proposes and confirms that 6-Shogaol can significantly inhibit the migration of colon cancer cells Caco2 and HCT116, and its mechanism may be produced by inhibiting EMT through IKKß/NF-κB/Snail signaling pathway. It was also confirmed that 6-Shogaol inhibited the proliferation and promoted apoptosis of Caco2 and HCT116 cells.


Assuntos
Neoplasias do Colo , NF-kappa B , Humanos , NF-kappa B/metabolismo , Quinase I-kappa B/farmacologia , Quinase I-kappa B/uso terapêutico , Metaloproteinase 2 da Matriz , Células CACO-2 , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Caderinas/metabolismo , Caderinas/farmacologia , Caderinas/uso terapêutico , Movimento Celular , Proliferação de Células , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal
3.
Cancer Biol Med ; 19(8)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35188360

RESUMO

OBJECTIVE: The novel compound GCJ-490A has been discovered as a pan-histone deacetylase (HDAC) inhibitor that exerts potent inhibitory activity against HDAC1, HDAC3, and HDAC6. Because of the important roles of HDACs in lung cancer development and the high distribution of GCJ-490A in lung tissue, we explored the anti-tumor potency of GCJ-490A against non-small cell lung cancer (NSCLC) in vitro and in vivo in this study. METHODS: The in vitro effects of GCJ-490A alone or combined with the EGFR inhibitor gefitinib against NSCLC were measured with proliferation, apoptosis, and colony formation assays. NSCLC xenograft models were used to investigate the efficacy of GCJ-490A combined with gefitinib for the treatment of NSCLC in vivo. Western blot assays, luciferase reporter assays, chromatin immunoprecipitation assays, quantitative real time-PCR, immunohistochemistry, and transcription factor activity assays were used to elucidate possible mechanisms. RESULTS: GCJ-490A effectively inhibited NSCLC cell proliferation and induced apoptosis in vitro and in vivo. Interestingly, inhibition of HDAC1 and HDAC6 by GCJ-490A increased histone acetylation at the IKKα promoter and enhanced IKKα transcription, thus decreasing c-Met. Moreover, this c-Met downregulation was found to be essential for the synergistic anti-tumor activity of GCJ-490A and gefitinib. CONCLUSIONS: These findings highlight the promising potential of HDAC inhibitors in NSCLC treatment and provide a rational basis for the application of HDAC inhibitors in combination with EGFR inhibitors in clinical trials.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Receptores ErbB/genética , Gefitinibe/farmacologia , Gefitinibe/uso terapêutico , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/uso terapêutico , Humanos , Quinase I-kappa B/metabolismo , Quinase I-kappa B/uso terapêutico , Neoplasias Pulmonares/patologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/uso terapêutico
4.
Pediatr Infect Dis J ; 41(5): 430-435, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35067643

RESUMO

BACKGROUND: Infants with inborn errors of immunity (IEI), born in countries where Bacillus-Calmette-Guerin (BCG) vaccination is recommended at birth, are at risk of developing infectious complications following vaccination. A prompt diagnosis of disseminated BCG infection in these infants is essential, as many will require stem cell transplantation (SCT) for the immunologic cure. In patients with IEI, the mortality risk from disseminated mycobacterial infection is high, both before and following SCT. METHODS: A 7-month-old Qatari infant with an IEI, homozygous IKBKB gene mutation, was evaluated at our institution for SCT. He had a history of recurrent pneumonias, but pretransplant evaluation revealed negative cultures from bronchoalveolar fluid, blood and urine. At 8 months of age, the infant developed skin nodules of unclear etiology, prompting additional evaluation. RESULTS: Given his profound immunosuppression and receipt of broad-spectrum antimicrobials, plasma metagenomic next-generation sequencing (mNGS) was obtained and identified Mycobacterium tuberculosis complex within 72 hours. A skin biopsy was performed, and antimycobacterial therapy was initiated. Mycobacterium bovis-BCG was confirmed from cultures 3 weeks later. Treatment was complicated by elevated serum liver transaminases and aminoglycoside-associated high-frequency hearing loss. The infant completed 14 months of treatment from engraftment. Evaluation for active BCG infection after SCT was negative. CONCLUSION: In an infant with a unique IEI, plasma mNGS provided the first diagnosis of disseminated BCG infection. We believe that early initiation of antimycobacterial treatment improved the infant's clinical outcome. Plasma mNGS testing should be considered as a noninvasive screen for infectious pathogens in children with IEIs before SCT.


Assuntos
Mycobacterium bovis , Tuberculose , Vacina BCG/efeitos adversos , Criança , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/uso terapêutico , Lactente , Recém-Nascido , Masculino , Mutação , Mycobacterium bovis/genética , Tuberculose/tratamento farmacológico
5.
Biomolecules ; 11(1)2020 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-33375283

RESUMO

The nuclear factor kappa B (NF-κB) is a ubiquitous transcription factor central to inflammation and various malignant diseases in humans. The regulation of NF-κB can be influenced by a myriad of post-translational modifications (PTMs), including phosphorylation, one of the most popular PTM formats in NF-κB signaling. The regulation by phosphorylation modification is not limited to NF-κB subunits, but it also encompasses the diverse regulators of NF-κB signaling. The differential site-specific phosphorylation of NF-κB itself or some NF-κB regulators can result in dysregulated NF-κB signaling, often culminating in events that induce cancer progression and other hyper NF-κB related diseases, such as inflammation, cardiovascular diseases, diabetes, as well as neurodegenerative diseases, etc. In this review, we discuss the regulatory role of phosphorylation in NF-κB signaling and the mechanisms through which they aid cancer progression. Additionally, we highlight some of the known and novel NF-κB regulators that are frequently subjected to phosphorylation. Finally, we provide some future perspectives in terms of drug development to target kinases that regulate NF-κB signaling for cancer therapeutic purposes.


Assuntos
Quinase I-kappa B/genética , NF-kappa B/genética , Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Quinase I-kappa B/uso terapêutico , NF-kappa B/uso terapêutico , Neoplasias/patologia , Neoplasias/terapia , Fosforilação/genética , Processamento de Proteína Pós-Traducional/genética , Transdução de Sinais/genética , Transcrição Gênica/genética
6.
J Pain ; 13(5): 485-97, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22564672

RESUMO

UNLABELLED: Inhibitor kappa B kinase (IKK)-mediated nuclear factor-kappa B (NF-κB) activation is a major pathway for transcriptional control of various pro-inflammatory factors. We here assessed whether activation of this pathway specifically in primary nociceptive neurons of the dorsal root ganglia (DRG) contributes to the development of nociceptive hypersensitivity. Mice carrying a cre-loxP-mediated deletion of inhibitor kappa B kinase beta (IKKß) in DRG neurons were protected from nerve injury-evoked allodynia and hyperalgesia. This effect was mimicked by systemic treatment with an IKKß inhibitor but was not observed upon specific inhibition of IKKß in the spinal cord, suggesting a specific role of IKKß in the peripheral neurons. The deletion of IKKß in DRG neurons did not affect constitutive neuronal NF-κB activity, but reduced nerve injury-evoked NF-κB stimulation in the DRG and was associated with reduced upregulation of interleukin-16, monocyte chemoattractant protein-1/chemokine (CC motif) ligand 2 (MCP-1/CCL2), and tumor necrosis factor alpha (TNFα) in the DRG. These cytokines evoked a rapid rise of intracellular calcium in subsets of primary DRG neurons. The results suggest that IKKß-mediated NF-κB stimulation in injured primary sensory neurons promotes cytokine and chemokine production and contributes thereby to the development of chronic pain. PERSPECTIVE: Inhibitors of IKK that do not pass the blood-brain barrier and act only in the periphery might be useful for reduction of the pro-inflammatory response in peripheral DRG neurons and reduce thereby nerve injury-evoked pain without affecting neuroprotective effects of NF-κB in the central nervous system.


Assuntos
Citocinas/metabolismo , Gânglios Espinais/patologia , Hiperalgesia/etiologia , Quinase I-kappa B/metabolismo , Nociceptores/metabolismo , Neuropatia Ciática/patologia , Regulação para Cima/fisiologia , Análise de Variância , Animais , Cálcio/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/farmacologia , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/genética , Quinase I-kappa B/deficiência , Quinase I-kappa B/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptores/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Neuropatia Ciática/complicações , Neuropatia Ciática/tratamento farmacológico , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
7.
Neurobiol Dis ; 43(3): 598-608, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21624467

RESUMO

The activation of nuclear factor κB (NF-κB) contributes to muscle degeneration that results from dystrophin deficiency in human Duchenne muscular dystrophy (DMD) and in the mdx mouse. In dystrophic muscle, NF-κB participates in inflammation and failure of muscle regeneration. Peptides containing the NF-κB Essential Modulator (NEMO) binding domain (NBD) disrupt the IκB kinase complex, thus blocking NF-κB activation. The NBD peptide, which is linked to a protein transduction domain to achieve in vivo peptide delivery to muscle tissue, was systemically delivered to mdx mice for 4 or 7 weeks to study NF-κB activation, histological changes in hind limb and diaphragm muscle and ex vivo function of diaphragm muscle. Decreased NF-κB activation, decreased necrosis and increased regeneration were observed in hind limb and diaphragm muscle in mdx mice treated systemically with NBD peptide, as compared to control mdx mice. NBD peptide treatment resulted in improved generation of specific force and greater resistance to lengthening activations in diaphragm muscle ex vivo. Together these data support the potential of NBD peptides for the treatment of DMD by modulating dystrophic pathways in muscle that are downstream of dystrophin deficiency.


Assuntos
Quinase I-kappa B/administração & dosagem , Quinase I-kappa B/farmacocinética , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular/farmacocinética , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Peptídeos/uso terapêutico , Animais , Diafragma/patologia , Diafragma/fisiologia , Modelos Animais de Doenças , Distrofina/deficiência , Distrofina/genética , Quinase I-kappa B/uso terapêutico , Peptídeos e Proteínas de Sinalização Intracelular/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Esquelético/inervação , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Necrose/prevenção & controle , Regeneração Nervosa/efeitos dos fármacos , Regeneração Nervosa/genética , Regeneração Nervosa/fisiologia , Peptídeos/administração & dosagem , Peptídeos/farmacocinética , Estrutura Terciária de Proteína/genética
8.
Mol Vis ; 16: 2502-10, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-21139997

RESUMO

PURPOSE: To investigate the efficacy and safety of cationic nano-copolymers CS-g-(PEI-b-mPEG) mediated IκB kinase beta (IKKß) targeting siRNA in modulating wound healing in a monkey model of glaucoma filtration surgery. METHODS: The IKKß targeting siRNAs were chemically synthesized and screened in cultured monkey Tenon's fibroblasts in vitro. Fourteen monkeys underwent trabeculectomy and were randomly allocated to one of three treatment regimens: subconjunctival injection of either CS-g-(PEI-b-mPEG)/IKKß-siRNA (six eyes, 50nM, at the time of surgery and 7 days post surgery) or phosphate buffered saline (four eyes), or treated with mitomycin C (MMC; four eyes, 0.2 mg/ml). Bleb survival and characteristics, and intraocular pressure, were evaluated over a 60-day period. Histology of the surgical eyes was performed to evaluate ocular scarring and fibrosis in each group. RESULTS: Subconjunctival injection of CS-g-(PEI-b-mPEG)/IKKß-siRNA was well tolerated in this model. Both siRNA and MMC significantly prolonged bleb survival compared with the PBS group (the medians for survival days were 45.5, 60, and 29.5 in the siRNA, MMC, and PBS groups, respectively, p<0.01). Higher blebs were observed in the siRNA group than in the PBS group (p<0.01), while the MMC group showed the highest blebs among three groups (p<0.01). The surgical eyes in both the siRNA and MMC groups had significantly larger bleb area compared with the PBS group (p<0.01), but there was no significant difference between the siRNA and MMC groups (p=0.214). There were no significant differences in IOP readings among the three groups on the designated days after surgery (all p>0.05). The histologic examination demonstrated that the eyes treated with siRNA showed a marked reduction in subconjunctival scar tissue compared with the eyes in the PBS group. The conjunctival epithelium appeared healthy without the acellularity that was present in the MMC group. CONCLUSIONS: Subconjunctival injection of cationic nano-copolymers mediated IKKß targeting siRNA is associated with improved surgical outcome in a monkey model of trabeculectomy. This novel approach may potentially be a more controlled alternative as an anti-scarring agent in glaucoma filtration surgery.


Assuntos
Cirurgia Filtrante , Glaucoma/cirurgia , Quinase I-kappa B/uso terapêutico , Nanopartículas/química , Polímeros/química , RNA Interferente Pequeno/metabolismo , Cicatrização , Animais , Bioensaio , Cátions , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Glaucoma/patologia , Glaucoma/fisiopatologia , Glaucoma/terapia , Haplorrinos , Quinase I-kappa B/genética , Mitomicina/farmacologia , Transfecção , Cicatrização/efeitos dos fármacos
9.
Neurochem Int ; 57(8): 876-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20868715

RESUMO

Inflammation plays an important role in ischemic pathology. NF-κB is a transcription factor that has a crucial role in inflammation and cell survival, but its precise role in the ischemic aftermath is still uncertain. Therefore, we evaluated the effect of intracerebroventricular administration of a highly specific NF-κB inhibitor peptide, IKK-NBD, on transient focal cerebral ischemic injury in rat using middle cerebral artery occlusion model. The assessment of ischemia-induced neurological deficits, alterations in the proinflammatory cytokine IL-1ß level, OX-42 immunoreactivity, changes in blood-brain barrier (BBB) permeability, reactive oxygen species (ROS) production and DNA fragmentation by terminal dUTP nick end labelling (TUNEL) were monitored at 24h post reperfusion following 1h of ischemia after pre-treatment with either 40µg of IKK-NBD or the inactive IKK-NBD peptide, which served as control. Pre-treatment with IKK-NBD peptide significantly ameliorated the cerebral ischemia-induced neurological deficits. Quantification of IL-1ß by ELISA revealed significantly reduced striatal IL-1ß level in IKK-NBD peptide treated rats. The treatment also resulted in reduced staining of microglial OX-42 and significantly reduced extravasation of Evans blue dye, indicating protection of BBB from ischemic insult. These results indicate that specific NF-κB inhibition downplays post-ischemic inflammation. Furthermore, reduction in DNA fragmentation as assessed by TUNEL staining also confirms the neuroprotective effect of IKK-NBD peptide. Thus, it may be inferred that IKK-NBD peptide reduces ischemic brain damage and this can, at least partly, be attributed to reduction in inflammation following ischemic injury.


Assuntos
Proteínas de Transporte/fisiologia , Quinase I-kappa B/fisiologia , Mediadores da Inflamação/fisiologia , Ataque Isquêmico Transitório/prevenção & controle , NF-kappa B/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/fisiologia , Peptídeos/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Proteínas de Transporte/uso terapêutico , Morte Celular/fisiologia , Quinase I-kappa B/uso terapêutico , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/prevenção & controle , Mediadores da Inflamação/uso terapêutico , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , NF-kappa B/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/uso terapêutico , Proteínas de Ligação a RNA , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA