Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 386, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38195694

RESUMO

Both lytic and apoptotic cell death remove senescent and damaged cells in living organisms. However, they elicit contrasting pro- and anti-inflammatory responses, respectively. The precise cellular mechanism that governs the choice between these two modes of death remains incompletely understood. Here we identify Gasdermin E (GSDME) as a master switch for neutrophil lytic pyroptotic death. The tightly regulated GSDME cleavage and activation in aging neutrophils are mediated by proteinase-3 and caspase-3, leading to pyroptosis. GSDME deficiency does not alter neutrophil overall survival rate; instead, it specifically precludes pyroptosis and skews neutrophil death towards apoptosis, thereby attenuating inflammatory responses due to augmented efferocytosis of apoptotic neutrophils by macrophages. In a clinically relevant acid-aspiration-induced lung injury model, neutrophil-specific deletion of GSDME reduces pulmonary inflammation, facilitates inflammation resolution, and alleviates lung injury. Thus, by controlling the mode of neutrophil death, GSDME dictates host inflammatory outcomes, providing a potential therapeutic target for infectious and inflammatory diseases.


Assuntos
Gasderminas , Lesão Pulmonar , Humanos , Neutrófilos , Apoptose , Piroptose
2.
J Vis Exp ; (195)2023 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-37246861

RESUMO

The average lifespan of a neutrophil is less than 24 h, which limits basic research on neutrophils and the application of neutrophil studies. Our previous research indicated that multiple pathways could mediate the spontaneous death of neutrophils. A cocktail was developed by simultaneously targeting these pathways, caspases-lysosomal membrane permeabilization-oxidant-necroptosis inhibition plus granulocyte colony-stimulating factor (CLON-G), which prolonged the neutrophil lifespan to greater than 5 days without significantly compromising the neutrophil function. Concurrently, a reliable and stable protocol for assessing and evaluating neutrophil death was also developed. In this work, we show that CLON-G can prolong the neutrophil lifespan in vitro to more than 5 days, and we exhibit the lengthening of the neutrophil lifespan with FACS and confocal fluorescence microscopy. This report introduces procedures for the preparation of CLON-G and showcases an in vitro spontaneous death assay of neutrophils, which can be used for the study of neutrophils and for subsequently interrogating neutrophil death, thus providing a reliable resource for the neutrophil community.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos , Neutrófilos , Neutrófilos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Longevidade , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Caspases/metabolismo
4.
Cancer Biol Med ; 19(12)2022 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-36514901

RESUMO

Neutrophils play an essential role in the defense against bacterial infections and orchestrate both the innate and adaptive immune responses. With their abundant numbers, diverse function and short life span, these cells are at the forefront of immune responses, and have gained attention in recent years because of their presence in tumor sites. Neutrophil involvement pertains to tumor cells' ability to construct a suitable tumor microenvironment (TME) that accelerates their own growth and malignancy, by facilitating their interaction with surrounding cells through the circulatory and lymphatic systems, thereby influencing tumor development and progression. Studies have indicated both pro- and anti-tumor properties of infiltrating neutrophils. The TME can exploit neutrophil function, recruitment, and even production, thus resulting in pro-tumor properties of neutrophils, including promotion of genetic instability, tumor cell proliferation, angiogenesis and suppression of anti-tumor or inflammatory response. In contrast, neutrophils can mediate anti-tumor resistance by direct cytotoxicity to the tumor cells or by facilitating anti-tumor functions via crosstalk with T cells. Here, we summarize current knowledge regarding the effects of neutrophil heterogeneity under homeostatic and tumor conditions, including neutrophil phenotype and function, in cancer biology.


Assuntos
Neoplasias , Neutrófilos , Humanos , Microambiente Tumoral , Neoplasias/patologia , Linfócitos T/patologia
5.
Cytotherapy ; 24(2): 172-182, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34426082

RESUMO

BACKGROUND AIMS: Hematopoietic stem and progenitor cells (HSPCs) are known to produce short-lived mature blood cells via proliferation and differentiation in a process that depends partially on regulatory cytokines from the bone marrow (BM) microenvironment. Delayed BM recovery after tremendous damage to the hematopoietic system can lead to neutropenia, anemia, thrombopenia and even death. However, efficiently promote BM recovery is still a big problem to be solved. Here, the authors aim to use heat-inactivated Escherichia coli (HIEC) to enhance BM recovery and further to find out the potential mechanism. METHODS: X-rad was used to establish HIEC/IL-1ß-induced radioprotection model. Single-cell RNA sequencing, RT-PCR, and western blotting were performed to detect the expression of IL-1R1 on HSPCs. Flow cytometry and automated hematology analyzer were used to analyze the percentage and absolute number of different populations of hematopoietic cells. The effects of IL-1ß on HSPCs were studied using in vivo and in vitro experiments. RESULTS: HIEC/IL-1ß pre-treatment can significantly increase the survival rate of lethally irradiated mice, and these mice showed better hematopoietic regeneration compared with control group. IL-1R was expressed on HSPCs, and IL-1ß could directly function on HSPCs to promote the proliferation and differentiation of HSPCs, and inhibit the apoptosis of HSPCs. CONCLUSIONS: HIEC pre-treatment can rescue lethally irradiated mice by promoting hematopoietic recovery via IL-1ß/IL-1R1 signaling, which can promote the proliferation of HSPCs by enhancing the cell cycle and attenuating the apoptosis of HSPCs.


Assuntos
Escherichia coli , Temperatura Alta , Animais , Medula Óssea , Diferenciação Celular , Células-Tronco Hematopoéticas , Camundongos
6.
Nat Commun ; 12(1): 6699, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795266

RESUMO

Candida albicans is the most common cause of fungal sepsis. Inhibition of inflammasome activity confers resistance to polymicrobial and LPS-induced sepsis; however, inflammasome signaling appears to protect against C. albicans infection, so inflammasome inhibitors are not clinically useful for candidiasis. Here we show disruption of GSDMD, a known inflammasome target and key pyroptotic cell death mediator, paradoxically alleviates candidiasis, improving outcomes and survival of Candida-infected mice. Mechanistically, C. albicans hijacked the canonical inflammasome-GSDMD axis-mediated pyroptosis to promote their escape from macrophages, deploying hyphae and candidalysin, a pore-forming toxin expressed by hyphae. GSDMD inhibition alleviated candidiasis by preventing C. albicans escape from macrophages while maintaining inflammasome-dependent but GSDMD-independent IL-1ß production for anti-fungal host defenses. This study demonstrates key functions for GSDMD in Candida's escape from host immunity in vitro and in vivo and suggests that GSDMD may be a potential therapeutic target in C. albicans-induced sepsis.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Inflamassomos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Macrófagos/imunologia , Proteínas de Ligação a Fosfato/imunologia , Animais , Candida albicans/fisiologia , Candidíase/genética , Candidíase/microbiologia , Caspase 1/genética , Caspase 1/imunologia , Caspase 1/metabolismo , Células Cultivadas , Feminino , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamassomos/genética , Inflamassomos/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Estimativa de Kaplan-Meier , Rim/imunologia , Rim/metabolismo , Rim/microbiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/metabolismo
7.
Sci Transl Med ; 13(604)2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321317

RESUMO

Clinical outcomes from granulocyte transfusion (GTX) are disadvantaged by the short shelf life and compromised function of donor neutrophils. Spontaneous neutrophil death is heterogeneous and mediated by multiple pathways. Leveraging mechanistic knowledge and pharmacological screening, we identified a combined treatment, caspases-lysosomal membrane permeabilization-oxidant-necroptosis inhibition plus granulocyte colony-stimulating factor (CLON-G), which altered neutrophil fate by simultaneously targeting multiple cell death pathways. CLON-G prolonged human and mouse neutrophil half-life in vitro from less than 1 day to greater than 5 days. CLON-G-treated aged neutrophils had equivalent morphology and function to fresh neutrophils, with no impairment to critical effector functions including phagocytosis, bacterial killing, chemotaxis, and reactive oxygen species production. Transfusion with stored CLON-G-treated 3-day-old neutrophils enhanced host defenses, alleviated infection-induced tissue damage, and prolonged survival as effectively as transfusion with fresh neutrophils in a clinically relevant murine GTX model of neutropenia-related bacterial pneumonia and systemic candidiasis. Last, CLON-G treatment prolonged the shelf life and preserved the function of apheresis-collected human GTX products both ex vivo and in vivo in immunodeficient mice. Thus, CLON-G treatment represents an effective and applicable clinical procedure for the storage and application of neutrophils in transfusion medicine, providing a therapeutic strategy for improving GTX efficacy.


Assuntos
Neutropenia , Neutrófilos , Idoso , Animais , Morte Celular , Fator Estimulador de Colônias de Granulócitos , Humanos , Transfusão de Leucócitos , Camundongos
8.
Nat Immunol ; 21(9): 1119-1133, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32719519

RESUMO

The full neutrophil heterogeneity and differentiation landscape remains incompletely characterized. Here, we profiled >25,000 differentiating and mature mouse neutrophils using single-cell RNA sequencing to provide a comprehensive transcriptional landscape of neutrophil maturation, function and fate decision in their steady state and during bacterial infection. Eight neutrophil populations were defined by distinct molecular signatures. The three mature peripheral blood neutrophil subsets arise from distinct maturing bone marrow neutrophil subsets. Driven by both known and uncharacterized transcription factors, neutrophils gradually acquire microbicidal capability as they traverse the transcriptional landscape, representing an evolved mechanism for fine-tuned regulation of an effective but balanced neutrophil response. Bacterial infection reprograms the genetic architecture of neutrophil populations, alters dynamic transitions between subpopulations and primes neutrophils for augmented functionality without affecting overall heterogeneity. In summary, these data establish a reference model and general framework for studying neutrophil-related disease mechanisms, biomarkers and therapeutic targets at single-cell resolution.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/fisiologia , Neutrófilos/fisiologia , Peritonite/imunologia , Análise de Célula Única/métodos , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Homeostase , Humanos , Camundongos , Análise de Sequência de RNA
9.
Front Immunol ; 11: 626, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373117

RESUMO

Pathogen-initiated chronic inflammation or autoimmune diseases accelerate proliferation and promote differentiation of hematopoietic stem cells (HSCs) but simultaneously reduce reconstitution capacity. Nevertheless, the effect of acute infection and inflammation on functional HSCs is still largely unknown. Here we found that acute infection elicited by heat-inactivated Escherichia coli (HIEC) expanded bone marrow lineage-negative (Lin)- stem-cell antigen 1 (Sca-1)+cKit+ (LSK) cell population, leading to reduced frequency of functional HSCs in LSK population. However, the total number of BM phenotypic HSCs (Flk2-CD48-CD150+ LSK cells) was not altered in HIEC-challenged mice. Additionally, the reconstitution capacity of the total BM between infected and uninfected mice was similar by both the competitive repopulation assay and measurement of functional HSCs by limiting dilution. Thus, occasionally occurring acute inflammation, which is critical for host defenses, is unlikely to affect HSC self-renewal and maintenance of long-term reconstitution capacity. During acute bacterial infection and inflammation, the hematopoietic system can replenish hematopoietic cells consumed in the innate inflammatory response by accelerating hematopoietic stem and progenitor cell proliferation, but preserving functional HSCs in the BM.


Assuntos
Células da Medula Óssea/fisiologia , Infecções por Escherichia coli/imunologia , Escherichia coli/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Inflamação/imunologia , Doença Aguda , Animais , Diferenciação Celular , Autorrenovação Celular , Células Cultivadas , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Quimeras de Transplante
10.
Curr Opin Hematol ; 26(1): 28-33, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30407218

RESUMO

PURPOSE OF REVIEW: CXCR2 is key stimulant of immune cell migration and recruitment, especially of neutrophils. Alleviating excessive neutrophil accumulation and infiltration could prevent prolonged tissue damage in inflammatory disorders. This review focuses on recent advances in our understanding of the role of CXCR2 in regulating neutrophil migration and the use of CXCR2 antagonists for therapeutic benefit in inflammatory disorders. RECENT FINDINGS: Recent studies have provided new insights into how CXCR2 signaling regulates hematopoietic cell mobilization and function in both health and disease. We also summarize several CXCR2 regulatory mechanisms during infection and inflammation such as via Wip1, T-bet, P-selectin glycoprotein ligand-1, granulocyte-colony-stimulating factor, and microbiome. Moreover, we provide an update of studies investigating CXCR2 blockade in the laboratory and in clinical trials. SUMMARY: Neutrophil homeostasis, migration, and recruitment must be precisely regulated. The CXCR2 signaling pathway is a potential target for modifying neutrophil dynamics in inflammatory disorders. We discuss the recent clinical use of CXCR2 antagonists for controlling inflammation.


Assuntos
Anti-Inflamatórios/uso terapêutico , Movimento Celular/efeitos dos fármacos , Microbiota/imunologia , Neutrófilos/imunologia , Receptores de Interleucina-8B , Animais , Movimento Celular/imunologia , Fator Estimulador de Colônias de Granulócitos/imunologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Glicoproteínas de Membrana/imunologia , Neutrófilos/patologia , Proteína Fosfatase 2C/imunologia , Receptores de Interleucina-8B/antagonistas & inibidores , Receptores de Interleucina-8B/imunologia , Proteínas com Domínio T/imunologia
11.
J Mol Cell Biol ; 11(6): 474-488, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30481289

RESUMO

Lin28a is a pluripotent factor that promotes somatic cell reprogramming. Unlike other pluripotent factors, Lin28a expression is transient and accumulated in primed embryonic stem (ES) cells, but its exact function and mechanism in the conversion of ES cells from naïve to primed state remain unclear. Here, we present evidence for Dppa3, a protein originally known for its role in germ cell development, as a downstream target of Lin28a in naïve-primed conversion. Using rescue experiment, we demonstrate that Dppa3 functions predominantly downstream of Lin28a during naïve-primed state conversion. Higher level of Lin28a prevents let-7 maturation and results in Dnmt3a/b (target of let-7) upregulation, which in turn induces hypermethylation of the Dppa3 promoter. Dppa3 demarcates naïve versus primed pluripotency states. These results emphasize that Lin28a plays an important role during the naïve-primed state conversion of ES cells, which is partially mediated by a Lin28a-let-7-Dnmt3a/b-Dppa3 axis.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Animais , Proteínas Cromossômicas não Histona/genética , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Regulação Enzimológica da Expressão Gênica , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Proteínas de Ligação a RNA/genética , Regulação para Cima , DNA Metiltransferase 3B
12.
J Cell Physiol ; 234(2): 1044-1051, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30256396

RESUMO

Signal transducer and activator of transcription 3 (Stat3) is a cytoplasmic transcription with many important functions, including regulation of cell proliferation, differentiation, survival, angiogenesis, and immune response. Besides, it plays critical roles in regulating the pluripotency. With the ability of self-renewal and differentiation, embryonic stem (ES) cells provide an unlimited source for cell transplantation. ES cells can maintain its undifferentiated state with leukemia inhibitory factor, the role which is achieved by the activation of the Stat3 pathway. Moreover, Stat3 activation is necessary for the naïve state maintenance of the ES cells and somatic stem cells reprogramming. This study presents an overview of the critical roles of Stat3 activation in the pluripotency maintenance of ES cells, somatic cell reprogramming, and naïve-primed pluripotent states conversion of ES cells.


Assuntos
Reprogramação Celular , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem da Célula , Técnicas de Reprogramação Celular , Humanos , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Camundongos , Fenótipo , Fator de Transcrição STAT3/genética , Transdução de Sinais
13.
Stem Cell Reports ; 11(5): 1092-1105, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30392974

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) undergo self-renewal and differentiation to guarantee a constant supply of short-lived blood cells. Both intrinsic and extrinsic factors determine HSPC fate, but the underlying mechanisms remain elusive. Here, we report that Proteinase 3 (PR3), a serine protease mainly confined to granulocytes, is also expressed in HSPCs. PR3 deficiency intrinsically suppressed cleavage and activation of caspase-3, leading to expansion of the bone marrow (BM) HSPC population due to decreased apoptosis. PR3-deficient HSPCs outcompete the long-term reconstitution potential of wild-type counterparts. Collectively, our results establish PR3 as a physiological regulator of HSPC numbers. PR3 inhibition is a potential therapeutic target to accelerate and increase the efficiency of BM reconstitution during transplantation.


Assuntos
Medula Óssea/enzimologia , Células-Tronco Hematopoéticas/enzimologia , Serina Endopeptidases/metabolismo , Animais , Apoptose , Medula Óssea/efeitos da radiação , Contagem de Células , Proliferação de Células , Sobrevivência Celular , Hematopoese , Células-Tronco Hematopoéticas/citologia , Camundongos Endogâmicos C57BL , Serina Endopeptidases/deficiência
14.
ACS Appl Mater Interfaces ; 10(36): 30081-30091, 2018 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-30118197

RESUMO

Mesenchymal stem cell (MSC)-derived exosomes have been recognized as new candidates for cell-free treatment of various diseases. However, maintaining the retention and stability of exosomes over time in vivo after transplantation is a major challenge in the clinical application of MSC-derived exosomes. Here, we investigated if human placenta-derived MSC-derived exosomes incorporated with chitosan hydrogel could boost the retention and stability of exosomes and further enhance their therapeutic effects. Our results demonstrated that chitosan hydrogel notably increased the stability of proteins and microRNAs in exosomes, as well as augmented the retention of exosomes in vivo as confirmed by Gaussia luciferase imaging. In addition, we assessed endothelium-protective and proangiogenesis abilities of hydrogel-incorporated exosomes in vitro. Meanwhile, we evaluated the therapeutic function of hydrogel-incorporated exosomes in a murine model of hindlimb ischemia. Our data demonstrated that chitosan hydrogel could enhance the retention and stability of exosomes and further augment the therapeutic effects for hindlimb ischemia as revealed by firefly luciferase imaging of angiogenesis. The strategy used in this study may facilitate the development of easy and effective approaches for assessing and enhancing the therapeutic effects of stem cell-derived exosomes.


Assuntos
Exossomos/transplante , Hidrogéis/química , Hidrogéis/farmacologia , Isquemia/terapia , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Exossomos/química , Membro Posterior/efeitos dos fármacos , Membro Posterior/patologia , Isquemia/tratamento farmacológico , Camundongos
15.
Cytotherapy ; 20(2): 181-188, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29269240

RESUMO

BACKGROUND AIMS: Imatinib (IM), a tyrosine kinase inhibitor targeting the BCR-ABL oncoprotein, remains a major therapeutic strategy for patients with chronic myelogenous leukemia (CML). However, IM resistance is still a challenge in the treatment of CML. Recently, it was reported that exosomes (Exo) were involved in drug resistance. Therefore, the present study investigated whether Exo secreted by human umbilical cord mesenchymal stromal cells (hUC-MSC-Exo) affected the sensitivity of K562 cells to IM. METHODS: hUC-MSC-Exo were isolated and identified. K562 cells were then treated or not with IM (1 µmol/L) in combination with hUC-MSC-Exo (50 µg/mL). Cell viability and apoptosis were determined by cell counting kit 8 (CCK-8) and annexin V/propidium iodide (PI) double staining, respectively. Apoptotic proteins, caspase and their cleaved forms were detected by Western blot. RESULTS: It was shown that hUC-MSC-Exo alone had no effect on cell viability and apoptosis of K562 cells. However, hUC-MSC-Exo promoted IM-induced cell viability inhibition and apoptosis. Moreover, hUC-MSC-Exo enhanced the increased Bax expression and the decreased Bcl-2 expression that were induced by IM. Compared with IM alone, caspase-9 and caspase-3 were further activated by combination of hUC-MSC-Exo with IM. Finally, the effects of hUC-MSC-Exo on K562 cells could be reversed by pretreatment of K562 cells with caspase inhibitor Z-VAD-FMK (30 µmol/L) DISCUSSION: These results indicate that hUC-MSC-Exo enhanced the sensitivity of K562 cells to IM via activation of caspase signaling pathway. Therefore, combining IM with hUC-MSC-Exo could be a promising approach to improve the efficacy of CML treatment.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Exossomos/metabolismo , Mesilato de Imatinib/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Sobrevivência Celular/efeitos dos fármacos , Exossomos/efeitos dos fármacos , Humanos , Células K562 , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
16.
Acta Biomater ; 63: 190-199, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28859902

RESUMO

Transplantation of endothelial cells (ECs) holds great promise for treating various kinds of ischemic diseases. However, the major challenge in ECs-based therapy in clinical applications is to provide high quality and enough amounts of cells. In this study, we developed a simple and efficient system to direct endothelial differentiation of mouse embryonic stem cells (ESCs) using a controllable chitosan nitric oxide (NO)-releasing hydrogel (CS-NO). ESCs were plated onto the hydrogel culture system, and the expressions of differentiation markers were measured. We found that the expression of Flk-1 (early ECs marker) and VE-cadherin (mature ECs marker) increased obviously under the controlled NO releasing environment. Moreover, the Flk-1 upregulation was accompanied by the activation of the phospho-inositide-3 kinase (PI3K)/Akt signaling. We also found that in the presence of the PI3K inhibitor (LY294002), the endothelial commitment of ESCs was abolished, indicating the importance of Akt phosphorylation in the endothelial differentiation of ESCs. Interestingly, in the absence of NO, the activation of Akt phosphorylation alone by using AKT activator (SC-79) did not profoundly promote the endothelial differentiation of ESCs, suggesting an interdependent relationship between NO and the Akt phosphorylation in driving endothelial fate specification of ESCs. Taken together, we demonstrated that NO releasing in a continuous and controlled manner is a simple and efficient method for directing the endothelial differentiation of ESCs without adding growth factors. STATEMENT OF SIGNIFICANCE: Fascinating data continues to show that artificial stem cell niche not only serve as a physical supporting scaffold for stem cells proliferation, but also as a novel platform for directing stem cell differentiation. Because of the lack of proper microenvironment for generating therapeutic endothelial cells (ECs) in vitro, the source of ECs for transplantation is the major limitation in ECs-based therapy to clinical applications. The current study established a feeder cell-free, 2-dimensional culture system for promoting the differentiation processes of embryonic stem cells (ESCs) committed to the endothelial lineage via using a nitric oxide (NO) controlled releasing hydrogel (CS-NO). Notably, the NO releasing from the hydrogel could selectively up-regulate Flk-1 (early ECs marker) and VE-cadherin (mature ECs marker) in the absence of growth factors, which was of crucial importance in the endothelial differentiation of ESCs. In summary, the current study proposes a simple and efficient method for directing the endothelial differentiation of ESCs without extra growth factors.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Endoteliais/citologia , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Células-Tronco Embrionárias Murinas/citologia , Óxido Nítrico/farmacologia , Animais , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
17.
Stem Cells Dev ; 26(12): 912-931, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28418782

RESUMO

Mesenchymal stem cells (MSCs) have shown promise for use in cell therapy, and due to their tumor tropism can serve as vehicles for delivering therapeutic agents to tumor sites. Because interleukin-8 (IL-8) is known to mediate the protumor effect of MSCs, elimination of IL-8 secretion by MSCs may enhance their safety for use in cancer gene therapy. However, little is known concerning the effect of endogenously secreted IL-8 on MSCs. We performed studies using placenta-derived MSCs (PMSCs) to determine whether knockdown of IL-8 would influence their biological activity. We first verified that IL-8 and its membrane receptor CXCR2, but not CXCR1, were highly expressed in PMSCs. We then employed lentivirus-mediated small hairpin RNA interference to generate stable IL-8-silenced PMSCs, which displayed a variety of characteristic senescent phenotypes. We observed that at day 9 post-transfection, IL-8-silenced PMSCs had become larger and displayed a more flattened appearance when compared with their controls. Moreover, their proliferation, colony forming unit-fibroblast formation, adipogenic and osteogenic differentiation, and immunosuppressive potentials were significantly impaired. Enhanced senescence-associated ß-galactosidase (SA-ß-gal) activity and specific global gene expression profiles confirmed that IL-8 silencing evoked the senescence process in PMSCs. Increased levels of p-Akt and decreased levels of FOXO3a protein expression suggested that reactive oxygen species played a role in the initiation and maintenance of senescence in IL-8-silenced PMSCs. Notably, the majority of CXCR2 ligands were downregulated in presenescent IL-8-silenced PMSCs but upregulated in senescent cells, indicating an antagonistic pleiotropy of the IL-8/CXCR2 signaling pathway in PMSCs. This effect may promote the proliferation of young cells and accelerate senescence of old cells.


Assuntos
Proliferação de Células/genética , Senescência Celular/genética , Técnicas de Silenciamento de Genes , Interleucina-8/genética , Células-Tronco Mesenquimais/metabolismo , Placenta/metabolismo , Feminino , Humanos , Interleucina-8/metabolismo , Células-Tronco Mesenquimais/citologia , Placenta/citologia , Gravidez , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo
18.
Mol Med Rep ; 14(5): 4187-4197, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27667024

RESUMO

Numerous studies have demonstrated that interferon-γ (IFN-γ) is an important inflammatory cytokine, which may activate the immunomodulatory abilities of mesenchymal stem cells (MSCs), and may influence certain other functions of these cells. MicroRNAs are small non­coding RNAs that regulate the majority of the biological functions of cells and are important in a variety of biological processes. However, few studies have been performed to investigate whether IFN­Î³ affects the microRNA profile of MSCs. The aim of the present study was to analyze the microRNA profile of MSCs derived from the umbilical cord (UC­MSCs) cultured in the presence or absence of IFN­Î³ (IFN­UC­MSCs). An array that detects 754 microRNAs was used to determine the expression profiles. Statistical analysis of the array data revealed that 8 microRNAs were significantly differentially expressed in UC­MSCs and IFN­UC­MSCs. Reverse transcription­quantitative polymerase chain reaction validated the differential expression of the 8 identified microRNAs. The target genes of the 8 microRNAs were predicted through two online databases, TargetScan and miRanda, and the predicted results were screened by bioinformatics analysis. The majority of the target genes were involved in the regulation of transcription, signal transduction, proliferation, differentiation and migration. These results may provide insight into the mechanism underlying the regulation of the biological functions of MSCs by IFN­Î³, in particular the immunomodulatory activity.


Assuntos
Interferon gama/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/biossíntese , Cordão Umbilical/metabolismo , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunomodulação/genética , Interferon gama/administração & dosagem , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , MicroRNAs/metabolismo , Cordão Umbilical/citologia
19.
Heart Surg Forum ; 19(3): E094-6, 2016 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-27355139

RESUMO

BACKGROUND: Few data were known on surgical management of intracardiac-extending in patients with intravenous leiomyomatosis (IVL). METHODS: From June 2007 to December 2014, six women (mean age, 39.3 ± 7.5 years; range, 24-55 years) with intracardiac-extending IVL were treated surgically at our hospital. Data were obtained from medical and pathological records, including characteristics of patients, surgical management, and follow-up. RESULTS: Surgery was performed successfully in all patients. Of 6 patients, 4 underwent one-stage operation and 2 underwent two-stage procedures. Circulatory arrest with hypothermia was used for a cardiotomy combined with venotomy in 5 patients. Complete resection was done in 5 patients. There were no perioperative deaths or complications in any of the patients. Hospital stay was 11.2 ± 2.9 days (range 7-15 days). All patients were followed-up for a mean of 41.0 ± 19.1 months (range, 17-69 months) after surgery. A recurrence of pelvic mass was found in 1 patient, but no symptoms or intravenous mass were reported. No obstruction occurred in any patient with a venotomy. CONCLUSION: Surgery is a better therapy for IVL and complete removal has favorable outcomes.


Assuntos
Leiomiomatose/cirurgia , Neoplasias Vasculares/cirurgia , Adulto , Feminino , Humanos , Tempo de Internação , Pessoa de Meia-Idade , Resultado do Tratamento
20.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 38(2): 164-8, 2016 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-27181892

RESUMO

OBJECTIVE: To investigate the effect of human umbilical cord-derived mesenchymal stem cells (UC-MSC) on the differentiation of leukemic cells. METHODS: The co-culture system of UC-MSC with acute promyelocytic leukemic cell line NB4 cells was constructed in vitro,and the differentiation status of the leukemic cells was assessed by cell morphology,nitroblue tetrazolium reduction test,and cell surface differentiation marker CD11b. RESULTS: UC-MSC induced the granulocytic differentiation of NB4 cells. When UC-MSC and a small dose of all-trans retinoic acid were applied together,the differentiation-inducing effect was enhanced in an additive manner. Interleukin (IL)-6Ra neutralization attenuated differentiation and exogenous IL-6-induced differentiation of leukemic cells. CONCLUSION: UC-MSC can promotd granulocytic differentiation of acute promyelocytic leukemia cells by way of IL-6 and presented additive effect when combined with a small dose of all-trans retinoic acid.


Assuntos
Diferenciação Celular , Interleucina-6/metabolismo , Leucemia Promielocítica Aguda/patologia , Células-Tronco Mesenquimais/metabolismo , Linhagem Celular Tumoral , Humanos , Tretinoína/farmacologia , Cordão Umbilical/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...