Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nature ; 621(7980): 830-839, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37674079

RESUMO

The immune-suppressive tumour microenvironment represents a major obstacle to effective immunotherapy1,2. Pathologically activated neutrophils, also known as polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), are a critical component of the tumour microenvironment and have crucial roles in tumour progression and therapy resistance2-4. Identification of the key molecules on PMN-MDSCs is required to selectively target these cells for tumour treatment. Here, we performed an in vivo CRISPR-Cas9 screen in a tumour mouse model and identified CD300ld as a top candidate of tumour-favouring receptors. CD300ld is specifically expressed in normal neutrophils and is upregulated in PMN-MDSCs upon tumour-bearing. CD300ld knockout inhibits the development of multiple tumour types in a PMN-MDSC-dependent manner. CD300ld is required for the recruitment of PMN-MDSCs into tumours and their function to suppress T cell activation. CD300ld acts via the STAT3-S100A8/A9 axis, and knockout of Cd300ld reverses the tumour immune-suppressive microenvironment. CD300ld is upregulated in human cancers and shows an unfavourable correlation with patient survival. Blocking CD300ld activity inhibits tumour development and has synergistic effects with anti-PD1. Our study identifies CD300ld as a critical immune suppressor present on PMN-MDSCs, being required for tumour immune resistance and providing a potential target for cancer immunotherapy.


Assuntos
Células Supressoras Mieloides , Neoplasias , Neutrófilos , Receptores Imunológicos , Animais , Humanos , Camundongos , Sistemas CRISPR-Cas , Progressão da Doença , Edição de Genes , Imunoterapia , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/patologia , Neoplasias/imunologia , Neoplasias/patologia , Neutrófilos/imunologia , Neutrófilos/patologia , Receptores Imunológicos/imunologia , Análise de Sobrevida , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/patologia , Microambiente Tumoral , Ativação Linfocitária
2.
Eur J Immunol ; 53(9): e2350374, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37417726

RESUMO

Atopic dermatitis (AD) is a common inflammatory skin disorder. Mast cells play an important role in AD because they regulate allergic reactions and inflammatory responses. However, whether and how the modulation of mast cell activity affects AD has not been determined. In this study, we aimed to determine the effects and mechanisms of 3-O-cyclohexanecarbonyl-11-keto-ß-boswellic acid (CKBA). This natural compound derivative alleviates skin inflammation by inhibiting mast cell activation and maintaining skin barrier homeostasis in AD. CKBA markedly reduced serum IgE levels and alleviated skin inflammation in calcipotriol (MC903)-induced AD mouse model. CKBA also restrained mast cell degranulation both in vitro and in vivo. RNA-seq analysis revealed that CKBA downregulated the extracellular signal-regulated kinase (ERK) signaling in BM-derived mast cells activated by anti-2,4-dinitrophenol/2,4-dinitrophenol-human serum albumin. We proved that CKBA suppressed mast cell activation via ERK signaling using the ERK activator (t-butyl hydroquinone) and inhibitor (selumetinib; AZD6244) in AD. Thus, CKBA suppressed mast cell activation in AD via the ERK signaling pathway and could be a therapeutic candidate drug for AD.


Assuntos
Dermatite Atópica , Camundongos , Humanos , Animais , Dermatite Atópica/tratamento farmacológico , Mastócitos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imunoglobulina E/metabolismo , Transdução de Sinais , Inflamação/metabolismo , Dinitrofenóis/metabolismo , Dinitrofenóis/farmacologia , Dinitrofenóis/uso terapêutico , Citocinas/metabolismo
3.
Invest Ophthalmol Vis Sci ; 64(7): 34, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37351878

RESUMO

Purpose: Photoreceptor (PR) death is the ultimate cause of irreversible vision loss in retinal detachment (RD). Although microglial infiltration in the subretinal space (SRS) was observed after RD, the molecular mechanism underlying microglial activation and the outcomes of infiltrating microglia remain unclear. We aimed to uncover the mechanism of initiation of microglial activation to help explore potential therapy to promote PR survival. Methods: An RD model was conducted by injecting sodium hyaluronate into SRS of C57BL/6J wild type mice. Adenosine triphosphate (ATP) was measured by a ATP Microplate Assay Kit. Bioinformatics analysis was used to evaluate the upregulated receptor relating to ATP binding in human datasets and mouse transcriptomes of RD. Expression of P2X7, its downstream signaling pathways, and microglial pyroptosis were confirmed by qPCR, WB, and immunofluorescence in vivo and in vitro. The cell viability of PR was measured by cell counting kit-8. Brilliant Blue G, a P2X7 antagonist, was subretinally or intraperitoneally injected to inhibit microglial activation in vivo and was applied for microglia cell line treatment in vitro. The decrease in microglial activation and pyroptosis was detected by immunofluorescence and WB. The protective effect on PR was measured by hematoxylin and eosin staining, TUNEL assay, and electroretinogram analysis. Results: The results showed that extracellular ATP released in the SRS after RD triggered P2X7 activation and attracted microglia. The downstream cascade of inflammasome activation induced by P2X7 activation contributed to microglial pyroptosis and then to PR death. ATP-activated microglia led to PR death in vitro. P2X7 blockade rescued PR morphologically and functionally by inhibiting microglial activation and pyroptosis. Conclusions: These results elucidate that ATP-induced P2X7-mediated microglial activation leads to microglial pyroptosis, contributing to PR death. Appropriate inhibition of microglial pyroptosis might serve as a pharmacotherapeutic strategy for decreasing PR death in RD.


Assuntos
Piroptose , Descolamento Retiniano , Camundongos , Humanos , Animais , Microglia/metabolismo , Descolamento Retiniano/metabolismo , Camundongos Endogâmicos C57BL , Trifosfato de Adenosina/metabolismo , Receptores Purinérgicos P2X7
4.
Nat Commun ; 14(1): 2004, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-37037861

RESUMO

Dermal fibroblasts and cutaneous nerves are important players in skin diseases, while their reciprocal roles during skin inflammation have not been characterized. Here we identify an inflammation-induced subset of papillary fibroblasts that promotes aberrant neurite outgrowth and psoriasiform skin inflammation by secreting the extracellular matrix protein tenascin-C (TNC). Single-cell analysis of fibroblast lineages reveals a Tnc+ papillary fibroblast subset with pro-axonogenesis and neuro-regulation transcriptomic hallmarks. TNC overexpression in fibroblasts boosts neurite outgrowth in co-cultured neurons, while fibroblast-specific TNC ablation suppresses hyperinnervation and alleviates skin inflammation in male mice modeling psoriasis. Dermal γδT cells, the main producers of type 17 pathogenic cytokines, frequently contact nerve fibers in mouse psoriasiform lesions and are likely modulated by postsynaptic signals. Overall, our results highlight the role of an inflammation-responsive fibroblast subset in facilitating neuro-immune synapse formation and suggest potential avenues for future therapeutic research.


Assuntos
Psoríase , Tenascina , Masculino , Camundongos , Animais , Tenascina/genética , Tenascina/metabolismo , Neuroimunomodulação , Proteínas da Matriz Extracelular/metabolismo , Modelos Animais de Doenças , Psoríase/metabolismo , Fibroblastos/metabolismo , Inflamação/patologia
5.
Immunotherapy ; 15(6): 429-442, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36880262

RESUMO

Aims: Immunotherapy has revolutionized cancer management. However, response to immunotherapy is heterogeneous. Thus, strategies to improve antitumor immune responses in resistant tumors, such as breast cancer, are urgently needed. Methods: Established murine tumors were treated with anti-CTLA4 or anti-PD-1 alone or combined with metronomic gemcitabine (met-GEM). Tumor vascular function, immune cell tumor infiltration and gene transcription were determined. Results: Low-dose met-GEM (2 mg/kg) treatments improved tumor vessel perfusion and increased tumor-infiltrating T cells. Notably, low-dose met-GEM pretreatments converted resistant tumors to respond to immunotherapy. Moreover, combined therapy reduced tumor vessel density, improved tumor vessel perfusion, increased T-cell tumor infiltration and upregulated the expression of some anticancer genes. Conclusion: Low-dose met-GEM pretreatment reconditioned the tumor immune microenvironment and improved immunotherapy efficacy in murine breast cancer.


Breast cancer is the most commonly diagnosed cancer in women globally. However, only a small subset of breast cancer patients benefits from treatment with immunotherapy. Thus, strategies aiming to enhance the antitumor effects of immunotherapies in breast cancer are an urgent requirement. We found that low-dose metronomic gemcitabine treatments improved tumor vessel function and increased tumor-infiltrating T cells, and did not deplete myeloid-derived suppressor cells, but did not affect tumor growth in the murine breast cancer model. Notably, however, low-dose metronomic gemcitabine pretreatments sensitized breast cancers to immunotherapy. Our findings provide important insights into the optimal strategies for combining immunotherapy with improved tumor vessel perfusion.


Assuntos
Neoplasias da Mama , Gencitabina , Humanos , Animais , Camundongos , Feminino , Imunoterapia , Linfócitos T , Linhagem Celular Tumoral , Microambiente Tumoral
6.
Membranes (Basel) ; 12(8)2022 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-36005661

RESUMO

This paper presented an innovative method for fabrication of a porous Al2O3/CaAl12O19 ceramic membrane by combining emulsion method, cement curing and tape-casting technologies. The ceramic membrane featured a smooth surface and a porous internal structure. By adjusting the oil-water volume ratio from 1:1 to 4:1, the porosity of the samples increases from 45.6 to 67.3%, density decreases from 2.07 to 1.32 g/cm3 and bending strength decreases from 64.3 ± 1.2 to 31.7 ± 0.6 MPa. More significantly, the membranes showed great gas permeability (1.2 × 107-2.3 × 107 Lm-2 h-1 bar-1), opening up a wide range of applications in the field of gas filtration processes.

7.
J Immunother Cancer ; 10(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35623658

RESUMO

BACKGROUND: Cancer vaccines are able to achieve tumor-specific immune editing in early-phase clinical trials. However, the infiltration of cytotoxic T cells into immune-deserted tumors is still a major limiting factor. An optimized vaccine approach to induce antigen-specific T cells that can perform robust tumor infiltration is important to accelerate their clinical translation. We previously developed a STING-activating PC7A nanovaccine that produces a strong anti-tumor T cell response on subcutaneous injection. This study systematically investigated the impact of administration methods on the performance of nanovaccines. METHODS: Tumor growth inhibition by intratumoral delivery and subcutaneous delivery of nanovaccine was investigated in TC-1 human papillomavirus-induced cancer model and B16-OVA melanoma model. Nanovaccine distribution in vivo was detected by clinical camera imaging, systemic T cell activation and tumor infiltration were tested by in vivo cytotoxicity killing assay and flow cytometry. For mechanism analysis, T cell recruitment was investigated by in vivo migration blocking assay, multiplex chemokine array, flow cytometry, RT-qPCR, chemotaxis assay and gene knockout mice. RESULTS: Nanovaccine administration was found to alter T cell production and infiltration in tumors. Intratumoral delivery of nanovaccines displayed superior antitumor effects in multiple tumor models compared with subcutaneous delivery. Mechanistic investigation revealed that intratumoral administration of the nanovaccine significantly increased the infiltration of antigen-specific T cells in TC-1 tumors, despite the lower systemic levels of T cells compared with subcutaneous injection. The inhibition of tumor growth by nanovaccines is primarily dependent on CD8+ cytotoxic T cells. Nanovaccine accumulation in tumors upregulates CXCL9 expression in myeloid cells in a STING dependent manner, leading to increased recruitment of IFNγ-expressing CD8+ T cells from the periphery, and IFNγ reciprocally stimulates CXCL9 expression in myeloid cells, resulting in positive feedback between myeloid-CXCL9 and T cell-IFNγ to promote T cell recruitment. However, the STING agonist alone could not sustain this effect in the presence of a systemic deficiency in antigen-specific T cells. CONCLUSIONS: Our results demonstrate that intratumoral administration of PC7A nanovaccine achieved stronger antitumor immunity and efficacy over subcutaneous injection. These data suggest intratumoral administration should be included in the therapeutic design in the clinical use of nanovaccine.


Assuntos
Vacinas Anticâncer , Melanoma Experimental , Nanopartículas , Animais , Linfócitos T CD8-Positivos , Humanos , Fatores Imunológicos/farmacologia , Imunoterapia/métodos , Melanoma Experimental/terapia , Camundongos
9.
Cell Res ; 32(1): 24-37, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34837059

RESUMO

Host cellular receptors play key roles in the determination of virus tropism and pathogenesis. However, little is known about SARS-CoV-2 host receptors with the exception of ACE2. Furthermore, ACE2 alone cannot explain the multi-organ tropism of SARS-CoV-2 nor the clinical differences between SARS-CoV-2 and SARS-CoV, suggesting the involvement of other receptor(s). Here, we performed genomic receptor profiling to screen 5054 human membrane proteins individually for interaction with the SARS-CoV-2 capsid spike (S) protein. Twelve proteins, including ACE2, ASGR1, and KREMEN1, were identified with diverse S-binding affinities and patterns. ASGR1 or KREMEN1 is sufficient for the entry of SARS-CoV-2 but not SARS-CoV in vitro and in vivo. SARS-CoV-2 utilizes distinct ACE2/ASGR1/KREMEN1 (ASK) receptor combinations to enter different cell types, and the expression of ASK together displays a markedly stronger correlation with virus susceptibility than that of any individual receptor at both the cell and tissue levels. The cocktail of ASK-related neutralizing antibodies provides the most substantial blockage of SARS-CoV-2 infection in human lung organoids when compared to individual antibodies. Our study revealed an interacting host receptome of SARS-CoV-2, and identified ASGR1 and KREMEN1 as alternative functional receptors that play essential roles in ACE2-independent virus entry, providing insight into SARS-CoV-2 tropism and pathogenesis, as well as a community resource and potential therapeutic strategies for further COVID-19 investigations.


Assuntos
COVID-19 , Glicoproteína da Espícula de Coronavírus , Receptor de Asialoglicoproteína , Recursos Comunitários , Humanos , Proteínas de Membrana , Ligação Proteica , Receptores Virais/metabolismo , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus
11.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34035167

RESUMO

The immunosuppressive and hypoxic tumor microenvironment (TME) remains a major obstacle to impede cancer immunotherapy. Here, we showed that elevated levels of Delta-like 1 (DLL1) in the breast and lung TME induced long-term tumor vascular normalization to alleviate tumor hypoxia and promoted the accumulation of interferon γ (IFN-γ)-expressing CD8+ T cells and the polarization of M1-like macrophages. Moreover, increased DLL1 levels in the TME sensitized anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA4) treatment in its resistant tumors, resulting in tumor regression and prolonged survival. Mechanically, in vivo depletion of CD8+ T cells or host IFN-γ deficiency reversed tumor growth inhibition and abrogated DLL1-induced tumor vascular normalization without affecting DLL1-mediated macrophage polarization. Together, these results demonstrate that elevated DLL1 levels in the TME promote durable tumor vascular normalization in a CD8+ T cell- and IFN-γ-dependent manner and potentiate anti-CTLA4 therapy. Our findings unveil DLL1 as a potential target to persistently normalize the TME to facilitate cancer immunotherapy.


Assuntos
Vasos Sanguíneos/patologia , Linfócitos T CD8-Positivos/imunologia , Proteínas de Ligação ao Cálcio/fisiologia , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Animais , Feminino , Células HEK293 , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Neoplasias/terapia , Microambiente Tumoral
12.
Int J Cancer ; 146(6): 1730-1740, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31840816

RESUMO

Immune checkpoint blockade (ICB) has shown long-term survival benefits, but only in a small fraction of cancer patients. Recent studies suggest that improved vessel perfusion by ICB positively correlates with its therapeutic outcomes. However, the underlying mechanism of such a process remains unclear. Here, we show that anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA4) treatment-induced tumor vessel normalization was accompanied by an increased infiltration of eosinophils into breast tumors. Eosinophil accumulation was positively correlated with the responsiveness of a breast tumor to anti-CTLA4 therapy. Depletion of eosinophils subsequently negated vessel normalization, reduced antitumor immunity and attenuated tumor growth inhibition by anti-CTLA4 therapy. Moreover, intratumoral accumulation of eosinophils relied on T lymphocytes and interferon γ production. Together, these results suggest that eosinophils partially mediate the antitumor effects of CTLA4 blockade through vascular remodeling. Our findings uncover an unidentified role of eosinophils in anti-CTLA4 therapy, providing a potential new target to improve ICB therapy and to predict its efficacy.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Antígeno CTLA-4/antagonistas & inibidores , Eosinófilos/efeitos dos fármacos , Eosinófilos/metabolismo , Neoplasias/etiologia , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Biomarcadores , Linhagem Celular Tumoral , Eosinófilos/imunologia , Feminino , Humanos , Imunidade , Imunomodulação/efeitos dos fármacos , Imunofenotipagem , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Depleção Linfocítica , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
13.
Sensors (Basel) ; 19(18)2019 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-31540211

RESUMO

This paper presents microelectromechanical system (MEMS)-based electrochemical seismometers with two pairs of electrodes integrated on one chip. Both theoretical analysis and numerical simulations were conducted to reveal the working principle of the proposed electrochemical seismometers, finding that flow holes distributed on cathodes rather than anodes can produce significantly higher sensitivities. The proposed electrochemical seismometers were fabricated based on conventional micromachined processes with high fabrication repeatability. Sensitivity measurements of the proposed seismometers and their commercial counterpart of CME6011 were conducted, indicating the sensitivities of the proposed seismometer with flow holes on cathodes were two orders higher than the counterpart with flow holes on anodes and one order higher than CME6011 at dominant frequencies. Measurements of random ground motions based on the proposed seismometer with flow holes on cathodes and CME6011 were conducted, producing comparable noise levels without observable ground motions and high correlations in response to random events of ground motions. These results validated the functionality of the proposed electrochemical seismometers, which may contribute to seismic monitoring in the near future.

14.
J Exp Clin Cancer Res ; 37(1): 260, 2018 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-30373628

RESUMO

BACKGROUND: Antiangiogenic agents are commonly used in lung and colon cancer treatments, however, rapid development of drug resistance limits their efficacy. METHODS: Lentinan (LNT) is a biologically active compound extracted from Lentinus edodes. The effects of LNT on tumor angiogenesis were evaluated by immunohistochemistry in murine LAP0297 lung and CT26 colorectal tumor models. The impacts of LNT on immune cells and gene expression in tumor tissues were determined by flow cytometry, qPCR, and ELISA. Nude mice and IFNγ blockade were used to investigate the mechanism of LNT affecting on tumor angiogenesis. The data sets were compared using two-tailed student's t tests or ANOVA. RESULTS: We found that LNT inhibited tumor angiogenesis and the growth of lung and colon cancers. LNT treatments elevated the expression of angiostatic factors such as IFNγ and also increased tumor infiltration of IFNγ-expressing T cells and myeloid cells. Interestingly, IFNγ blockade, but not T cell deficiency, reversed the effects of LNT treatments on tumor blood vessels. Moreover, long-lasting LNT administration persistently suppressed tumor angiogenesis and inhibited tumor growth. CONCLUSIONS: LNT inhibits tumor angiogenesis by increasing IFNγ production and in a T cell-independent manner. Our findings suggest that LNT could be developed as a new antiangiogenic agent for long-term treatment of lung and colon cancers.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Interferon gama/metabolismo , Lentinano/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Feminino , Interferon gama/genética , Lentinano/farmacologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Clin Invest ; 128(5): 2104-2115, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29664018

RESUMO

Immune checkpoint blockade (ICB) has demonstrated curative potential in several types of cancer, but only for a small number of patients. Thus, the identification of reliable and noninvasive biomarkers for predicting ICB responsiveness is an urgent unmet need. Here, we show that ICB increased tumor vessel perfusion in treatment-sensitive EO771 and MMTV-PyVT breast tumor as well as CT26 and MCA38 colon tumor models, but not in treatment-resistant MCaP0008 and 4T1 breast tumor models. In the sensitive tumor models, the ability of anti-cytotoxic T lymphocyte-associated protein 4 or anti-programmed cell death 1 therapy to increase vessel perfusion strongly correlated with its antitumor efficacy. Moreover, globally enhanced tumor vessel perfusion could be detected by Doppler ultrasonography before changes in tumor size, which predicted final therapeutic efficacy with more than 90% sensitivity and specificity. Mechanistically, CD8+ T cell depletion, IFN-γ neutralization, or implantation of tumors in IFN-γ receptor knockout mice abrogated the vessel perfusion enhancement and antitumor effects of ICB. These results demonstrated that ICB increased vessel perfusion by promoting CD8+ T cell accumulation and IFN-γ production, indicating that increased vessel perfusion reflects the successful activation of antitumor T cell immunity by ICB. Our findings suggest that vessel perfusion can be used as a novel noninvasive indicator for predicting ICB responsiveness.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo , Depleção Linfocítica , Neoplasias Mamárias Experimentais , Perfusão , Animais , Linfócitos T CD8-Positivos/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Interferon gama/genética , Interferon gama/metabolismo , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Camundongos
16.
Biochem Biophys Res Commun ; 450(4): 1261-5, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-24984152

RESUMO

OBJECTIVE: Recently we have reported that cleaved high molecular weight kininogen (HKa) accelerates the onset of endothelial progenitor cells (EPCs) senescence by induction of reactive oxygen species (ROS). However, the mechanisms by which HKa induces production of ROS remain unknown. In this study, we have shown that HKa induces EPC senescence via stimulation of c-Jun N-terminal kinases (JNK)-related pathway. METHODS AND RESULTS: Treatment of human EPCs with HKa for 72h stimulated JNK phosphorylation at Thr183/Tyr185, and FOXO4 phosphorylation at Thr451, Concomitantly, upregulated the expression of MnSOD at protein and mRNA levels in a concentration-dependent manner. HKa increased intracellular level of H2O2, without affecting the expression of catalase. To narrow down the functional domain of HKa, recombinant proteins of human HK heavy chain (HC, 19-380aa) and light chain (LC, 390-644aa) were generated. HC, but not LC, increased senescence of EPCs and intracellular ROS levels, to a similar extent with HKa. Moreover, HC at 50 nM increased FOXO4 phosphorylation at Thr451 and the protein level of MnSOD in EPCs. CONCLUSION: These results demonstrate that HKa accelerates the onset of EPC senescence by stimulating JNK/FOXO4/MnSOD pathway, its effect is mediated by the HC.


Assuntos
Senescência Celular , Endotélio/citologia , Cininogênios/metabolismo , MAP Quinase Quinase 4/metabolismo , Células-Tronco/citologia , Superóxido Dismutase/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Bases , Proteínas de Ciclo Celular , Células Cultivadas , Primers do DNA , Endotélio/enzimologia , Endotélio/metabolismo , Fatores de Transcrição Forkhead , Humanos , Cininogênios/química , Peso Molecular , Células-Tronco/enzimologia , Células-Tronco/metabolismo
17.
Radiat Res ; 177(6): 792-803, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22175298

RESUMO

L-arginine is shown to protect hematopoietic progenitor (32D cl 3) cells from death due to exposure to γ radiation ((137)Cs). Some of the other intermediates in the urea cycle, namely ornithine and citrulline, plus urea itself, were not found to have any significant impact on cell survival after irradiation. Intriguingly, supplementation of irradiated cells with L-arginine results in decreased production of peroxynitrite, suggesting that suppression of superoxide generation by nitric oxide synthase in one or more microenvironments is an important factor in the observed radioprotection. The absence of any radioprotective effect of L-arginine in cells at 3% oxygen also confirms the involvement of one or more oxygen-derived species. Knockdown experiments with nitric oxide synthase (NOS) siRNAs in cells and NOS knockout animals confirm that the observed radioprotection is associated with nNOS (NOS-1). L-arginine also ameliorates the transient inhibition of the electron-transport chain complex I that occurs within 30 min of completing the dose (10 Gy) and that appears to be a functional marker for postirradiation mitochondrial oxidant production.


Assuntos
Arginina/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos da radiação , Protetores contra Radiação/farmacologia , Animais , Benzimidazóis/metabolismo , Carbocianinas/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/efeitos da radiação , Miocárdio/citologia , Óxido Nítrico/biossíntese , Oxidantes/biossíntese , Oxidantes/metabolismo , Ácido Peroxinitroso/biossíntese , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA