Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Front Immunol ; 15: 1368099, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38665923

RESUMO

Early increase in the level of endothelial progenitor cells (EPCs) in the systemic circulation occurs in patients with septic infection/sepsis. The significance and underlying mechanisms of this response remain unclear. This study investigated the bone marrow EPC response in adult mice with septic infection induced by intravenous injection (i.v.) of Escherichia coli. For in vitro experiments, sorted marrow stem/progenitor cells (SPCs) including lineage(lin)-stem cell factor receptor (c-kit)+stem cell antigen-1 (Sca-1)-, lin-c-kit+, and lin- cells were cultured with or without lipopolysaccharides (LPSs) and recombinant murine vascular endothelial growth factor (VEGF) in the absence and presence of anti-Sca-1 crosslinking antibodies. In a separate set of experiments, marrow lin-c-kit+ cells from green fluorescence protein (GFP)+ mice, i.v. challenged with heat-inactivated E. coli or saline for 24 h, were subcutaneously implanted in Matrigel plugs for 5 weeks. Marrow lin-c-kit+ cells from Sca-1 knockout (KO) mice challenged with heat-inactivated E. coli for 24 h were cultured in the Matrigel medium for 8 weeks. The marrow pool of EPCs bearing the lin-c-kit+Sca-1+VEGF receptor 2 (VEGFR2)+ (LKS VEGFR2+) and LKS CD133+VEGFR2+ surface markers expanded rapidly following septic infection, which was supported by both proliferative activation and phenotypic conversion of marrow stem/progenitor cells. Increase in marrow EPCs and their reprogramming for enhancing angiogenic activity correlated with cell-marked upregulation of Sca-1 expression. Sca-1 was coupled with Ras-related C3 botulinum toxin substrate 2 (Rac2) in signaling the marrow EPC response. Septic infection caused a substantial increase in plasma levels of IFN-γ, VEGF, G-CSF, and SDF-1. The early increase in circulating EPCs was accompanied by their active homing and incorporation into pulmonary microvasculature. These results demonstrate that the marrow EPC response is a critical component of the host defense system. Sca-1 signaling plays a pivotal role in the regulation of EPC response in mice with septic infection.


Assuntos
Células Progenitoras Endoteliais , Proteínas de Membrana , Sepse , Animais , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/imunologia , Sepse/imunologia , Sepse/metabolismo , Camundongos , Camundongos Knockout , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Camundongos Endogâmicos C57BL , Fator A de Crescimento do Endotélio Vascular/metabolismo , Antígenos Ly/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/imunologia , Células Cultivadas , Masculino
2.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35163466

RESUMO

Ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury (AKI) and progression to chronic kidney disease (CKD). However, no effective therapeutic intervention has been established for ischemic AKI. Endothelial progenitor cells (EPCs) have major roles in the maintenance of vascular integrity and the repair of endothelial damage; they also serve as therapeutic agents in various kidney diseases. Thus, we examined whether EPCs have a renoprotective effect in an IRI mouse model. Mice were assigned to sham, EPC, IRI-only, and EPC-treated IRI groups. EPCs originating from human peripheral blood were cultured. The EPCs were administered 5 min before reperfusion, and all mice were killed 72 h after IRI. Blood urea nitrogen, serum creatinine, and tissue injury were significantly increased in IRI mice; EPCs significantly improved the manifestations of IRI. Apoptotic cell death and oxidative stress were significantly reduced in EPC-treated IRI mice. Administration of EPCs decreased the expression levels of NLRP3, cleaved caspase-1, p-NF-κB, and p-p38. Furthermore, the expression levels of F4/80, ICAM-1, RORγt, and IL-17RA were significantly reduced in EPC-treated IRI mice. Finally, the levels of EMT-associated factors (TGF-ß, α-SMA, Snail, and Twist) were significantly reduced in EPC-treated IRI mice. This study shows that inflammasome-mediated inflammation accompanied by immune modulation and fibrosis is a potential target of EPCs as a treatment for IRI-induced AKI and the prevention of progression to CKD.


Assuntos
Injúria Renal Aguda/prevenção & controle , Células Progenitoras Endoteliais/transplante , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Injúria Renal Aguda/metabolismo , Animais , Apoptose/efeitos dos fármacos , Nitrogênio da Ureia Sanguínea , Células Cultivadas , Creatinina/sangue , Modelos Animais de Doenças , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/imunologia , Células Progenitoras Endoteliais/metabolismo , Humanos , Masculino , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo
3.
Int J Mol Sci ; 22(24)2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34948469

RESUMO

Rheumatoid Arthritis (RA) is a chronic autoimmune inflammatory disease characterized by the swelling of multiple joints, pain and stiffness, and accelerated atherosclerosis. Sustained immune response and chronic inflammation, which characterize RA, may induce endothelial activation, damage and dysfunction. An equilibrium between endothelial damage and repair, together with the preservation of endothelial integrity, is of crucial importance for the homeostasis of endothelium. Endothelial Progenitor Cells (EPCs) represent a heterogenous cell population, characterized by the ability to differentiate into mature endothelial cells (ECs), which contribute to vascular homeostasis, neovascularization and endothelial repair. A modification of the number and function of EPCs has been described in numerous chronic inflammatory and auto-immune conditions; however, reports that focus on the number and functions of EPCs in RA are characterized by conflicting results, and discrepancies exist among different studies. In the present review, the authors describe EPCs' role and response to RA-related endothelial modification, with the aim of illustrating current evidence regarding the level of EPCs and their function in this disease, to summarize EPCs' role as a biomarker in cardiovascular comorbidities related to RA, and finally, to discuss the modulation of EPCs secondary to RA therapy.


Assuntos
Artrite Reumatoide/imunologia , Células Progenitoras Endoteliais/imunologia , Artrite Reumatoide/terapia , Biomarcadores/metabolismo , Regulação da Expressão Gênica , Humanos , Transdução de Sinais
4.
Microvasc Res ; 138: 104189, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34062191

RESUMO

Tumor-associated vessels constitution is the result of angiogenesis, the hallmark of cancer essential for tumor to develop in dimension and to spread throughout the organism. Tumor endothelium is configured as an active functioning organ capable of determine interaction with the immune response and all the other components of the variegate cancer microenvironment, determining reciprocal influence. Angiogenesis is here analyzed in its molecular and cellular mechanisms, multiple mediators and principal players, represented by Endothelial Cells. It is discussed the striking heterogeneity of cancer endothelium, due to morphological and molecular aberrations that it often presents and its multiple origin. Among the cells that participate to the composition of tumor vasculature, Endothelial Progenitor Cells represent an important source for physical sustain and paracrine signaling in the process of angiogenesis. Treatment options are reviewed, with particular focus on novel therapeutic strategies for overcoming tumor resistance to anti-angiogenic agents.


Assuntos
Células Progenitoras Endoteliais/patologia , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Microambiente Tumoral , Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Linhagem da Célula , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/imunologia , Células Progenitoras Endoteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/imunologia , Fenótipo , Transdução de Sinais , Microambiente Tumoral/imunologia
6.
Cell Commun Signal ; 19(1): 1, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33397378

RESUMO

BACKGROUND: Bone marrow derived endothelial progenitor cells (EPCs) are immature endothelial cells (ECs) involved in neo-angiogenesis and endothelial homeostasis and are considered as a circulating reservoir for endothelial repair. Many studies showed that EPCs from patients with cardiovascular pathologies are impaired and insufficient; hence, allogenic sources of EPCs from adult or cord blood are considered as good choices for cell therapy applications. However, allogenic condition increases the chance of immune rejection, especially by T cells, before exerting the desired regenerative functions. TNFα is one of the main mediators of EPC activation that recognizes two distinct receptors, TNFR1 and TNFR2. We have recently reported that human EPCs are immunosuppressive and this effect was TNFα-TNFR2 dependent. Here, we aimed to investigate if an adequate TNFα pre-conditioning could increase TNFR2 expression and prime EPCs towards more immunoregulatory functions. METHODS: EPCs were pre-treated with several doses of TNFα to find the proper dose to up-regulate TNFR2 while keeping the TNFR1 expression stable. Then, co-cultures of human EPCs and human T cells were performed to assess whether TNFα priming would increase EPC immunosuppressive and immunomodulatory effect. RESULTS: Treating EPCs with 1 ng/ml TNFα significantly up-regulated TNFR2 expression without unrestrained increase of TNFR1 and other endothelial injury markers. Moreover, TNFα priming through its interaction with TNFR2 remarkably enhanced EPC immunosuppressive and anti-inflammatory effects. Conversely, blocking TNFR2 using anti-TNFR2 mAb followed by 1 ng/ml of TNFα treatment led to the TNFα-TNFR1 interaction and polarized EPCs towards pro-inflammatory and immunogenic functions. CONCLUSIONS: We report for the first time the crucial impact of inflammation notably the TNFα-TNFR signaling pathway on EPC immunological function. Our work unveils the pro-inflammatory role of the TNFα-TNFR1 axis and, inversely the anti-inflammatory implication of the TNFα-TNFR2 axis in EPC immunoregulatory functions. Priming EPCs with 1 ng/ml of TNFα prior to their administration could boost them toward a more immunosuppressive phenotype. This could potentially lead to EPCs' longer presence in vivo after their allogenic administration resulting in their better contribution to angiogenesis and vascular regeneration. Video Abstract.


Assuntos
Células Progenitoras Endoteliais/efeitos dos fármacos , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Células Progenitoras Endoteliais/imunologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Imunomodulação , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
7.
Front Immunol ; 12: 798211, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975909

RESUMO

The bones and joints in the skeletal system are composed of diverse cell types, including vascular niches, bone cells, connective tissue cells and mineral deposits and regulate whole-body homeostasis. The capacity of maintaining strength and generation of blood lineages lies within the skeletal system. Bone harbours blood and immune cells and their progenitors, and vascular cells provide several immune cell type niches. Blood vessels in bone are phenotypically and functionally diverse, with distinct capillary subtypes exhibiting striking changes with age. The bone vasculature has a special impact on osteogenesis and haematopoiesis, and dysregulation of the vasculature is associated with diverse blood and bone diseases. Ageing is associated with perturbed haematopoiesis, loss of osteogenesis, increased adipogenesis and diminished immune response and immune cell production. Endothelial and perivascular cells impact immune cell production and play a crucial role during inflammation. Here, we discuss normal and maladapted vascular niches in bone during development, homeostasis, ageing and bone diseases such as rheumatoid arthritis and osteoarthritis. Further, we discuss the role of vascular niches during bone malignancy.


Assuntos
Envelhecimento/imunologia , Vasos Sanguíneos/imunologia , Doenças Ósseas/imunologia , Osso e Ossos/irrigação sanguínea , Células-Tronco Hematopoéticas/imunologia , Articulações/irrigação sanguínea , Nicho de Células-Tronco , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Diferenciação Celular , Proliferação de Células , Células Progenitoras Endoteliais/imunologia , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Homeostase , Humanos , Osteoartrite/imunologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Fenótipo
8.
Pediatr Res ; 90(1): 156-165, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33038874

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) ensure vascular integrity and neovascularization. No studies have investigated EPCs in preterm-born children beyond infancy. METHODS: One hundred and thirty-six prepubertal children were enrolled: 63 preterm and 73 born at term (controls). Circulating CD34(+)/VEGFR-2(+)/CD45(-) and CD34(+)/VEGFR-2(+)/CD45dim EPCs were measured in preterm-born children compared to controls. Body mass index (BMI), waist-to-hip ratio (WHR), neck circumference, systolic and diastolic blood pressure (SBP and DBP, respectively), fasting glucose, insulin, lipid profile, common carotid and abdominal aortic intima-media thickness (cIMT and aIMT, respectively), endothelium-dependent brachial artery flow-mediated dilation (FMD), and echocardiographic parameters were also assessed. RESULTS: Circulating CD34(+)/VEGFR-2(+)/CD45(-) and CD34(+)/VEGFR-2(+)/CD45dim EPCs were significantly higher in preterm-born children compared to controls (p < 0.001 and p < 0.001, respectively). In total study population and in the preterm-born group, EPCs were significantly lower in children born to mothers with gestational diabetes compared to non-diabetic mothers. Prematurity was associated with higher WHR, neck circumference, SBP, DBP, cIMT, aIMT, mean pressure, and velocity of pulmonary artery; the peak velocity of the brachial artery was significantly lower in children born prematurely. In multiple regression analysis, preterm birth and maternal gestational diabetes were recognized as independent predictors of EPCs. CONCLUSIONS: Circulating EPCs were increased in prepubertal preterm-born children in comparison with peers born full-term. Maternal gestational diabetes was associated with a decrease in EPCs. IMPACT: Mounting evidence supports the adverse effect of prematurity on cardiovascular health. However, the underlying mechanisms that could lead to endothelial dysfunction in preterm-born individuals are not fully understood. Endothelial progenitor cells (EPCs) ensure vascular integrity, normal endothelial function and neovascularization. No studies have investigated the EPCs counts in peripheral blood beyond infancy in children born prematurely. Circulating EPCs were significantly higher in preterm-born prepubertal children compared to controls, thus indicating that prematurity is possibly associated with endothelial damage. In total study population and in the preterm-born group, maternal gestational diabetes was associated with decreased EPCs concentrations.


Assuntos
Células Progenitoras Endoteliais/citologia , Fatores de Risco de Doenças Cardíacas , Nascimento Prematuro/fisiopatologia , Antígenos CD34/sangue , Artéria Braquial/fisiopatologia , Artérias Carótidas/fisiopatologia , Estudos de Casos e Controles , Criança , Células Progenitoras Endoteliais/imunologia , Feminino , Humanos , Antígenos Comuns de Leucócito/sangue , Masculino , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/sangue , Relação Cintura-Quadril
9.
Front Immunol ; 11: 581385, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33262760

RESUMO

Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.


Assuntos
Aterosclerose/etiologia , Aterosclerose/imunologia , Células Progenitoras Endoteliais/imunologia , Interferon Tipo I/imunologia , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Aterosclerose/fisiopatologia , Autoanticorpos/biossíntese , Linfócitos B/imunologia , Citocinas/imunologia , Células Progenitoras Endoteliais/classificação , Células Progenitoras Endoteliais/fisiologia , Armadilhas Extracelulares/imunologia , Humanos , Lúpus Eritematoso Sistêmico/fisiopatologia , Modelos Imunológicos , Estresse Oxidativo , Transdução de Sinais/imunologia , Linfócitos T/imunologia
10.
BMC Pulm Med ; 20(1): 306, 2020 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-33218322

RESUMO

BACKGROUND: Sarcoidosis is a multisystemic granulomatous disease with still unknown etiology. Our previous studies showed a significantly higher percentage of CD34 + cells in the peripheral blood in patients with sarcoidosis (SA) compared to the control group. The objective of the present study was to characterized of the CD34 + cell population in peripheral blood in patients with SA with reference to the control group. Moreover in patients with SA, fibrocytes and endothelial cells were analysed and their relationship to the fibrosis process based on assessment of diffusing capacity for carbon monoxide (DLCO). METHODS: Data from patients diagnosed with SA at Military Institute of Medicine (Warsaw, Poland) between January 2018 and December 2019 were collected and analysed ongoing basis. Peripheral blood was collected from 26 patients with newly diagnosed pulmonary SA and 16 healthy subjects. The immunomagnetic method and flow cytometry were used. Among the CD34+ progenitor cells were assessed: low-differentiated cells, hematopoietic progenitor cells and endothelial progenitor cells. The Statistica 12.0 software was used for a statistical analysis. RESULTS: We observed a significantly higher percentage of low-differentiated cells (13.8 vs. 2.3, P = 0.001) and endothelial cells (0.3 vs. 0.0, P = 0.001) in patients with SA compared to the control group. In the study group the median proportion of fibrocytes was 1.877% (0.983-2.340) in patients with DLCO< 80%, while in patients with DLCO> 80% was 0.795% (0.139-1.951) (P = 0.72). The median proportion of endothelial progenitor cells was higher in patients with DLCO< 80%: 0.889% (0.391-1.741), than in patients with DLCO> 80%: 0.451% (0.177-0.857) (P = 0.44). CONCLUSIONS: In conclusion we demonstrated for the first time the immunophenotype of peripheral CD34 + cells with the degree of their differentiation. The study confirmed the involvement of low differentiated cells and endothelial cells in patients with SA.


Assuntos
Antígenos CD34/imunologia , Células Progenitoras Endoteliais/imunologia , Sarcoidose Pulmonar/sangue , Sarcoidose Pulmonar/diagnóstico , Adulto , Idoso , Estudos de Casos e Controles , Diferenciação Celular , Progressão da Doença , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Polônia , Sarcoidose Pulmonar/imunologia
11.
J Autoimmun ; 112: 102486, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32482487

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune multisystem disease featured by an increased cardiovascular risk that may lead to premature patient's death. It has been demonstrated that SLE patients suffer from early onset endothelial dysfunction which is due to the impairment of endogenous vascular repair mechanisms. Vascular integrity and homeostasis are maintained by endothelial progenitor cells (EPCs), which are mobilized in response to endothelial injury to replace damaged endothelial cells. Two main EPCs subpopulations exist in peripheral blood: endothelial colony forming cells (ECFCs), which represent truly endothelial precursors and can physically engraft within neovessels, and myeloid angiogenic cells (MACs), which sustain angiogenesis in a paracrine manner. Emerging evidence indicates that ECFCs/MACs are down-regulated and display compromised angiogenic activity in SLE, thereby contributing to the pathogenesis of this disease. Intracellular calcium (Ca2+) signaling plays a crucial role in maintaining vascular integrity by stimulating migration, proliferation and tube formation in both ECFCs and MACs. Herein, we illustrate the evidences that support the role played by EPCs dysfunction in SLE. Subsequently, we discuss about the hypothesis that the Ca2+ handling machinery is compromised in SLE-derived ECFCs and MACs, thereby resulting in their reduced pro-angiogenic activity. Finally, we speculate about the proposal to exploit intracellular Ca2+ signaling to improve ECFCs' reparative phenotype and suggest this strategy as a new approach to treat SLE patients.


Assuntos
Sinalização do Cálcio/imunologia , Células Progenitoras Endoteliais/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Células Mieloides/metabolismo , Neovascularização Patológica/imunologia , Animais , Cálcio/metabolismo , Movimento Celular/imunologia , Proliferação de Células , Modelos Animais de Doenças , Células Progenitoras Endoteliais/imunologia , Humanos , Lúpus Eritematoso Sistêmico/patologia , Células Mieloides/imunologia , Neovascularização Patológica/patologia , Comunicação Parácrina/imunologia , Transdução de Sinais/imunologia
12.
Cell Commun Signal ; 18(1): 94, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32546175

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) are non-differentiated endothelial cells (ECs) present in blood circulation that are involved in neo-vascularization and correction of damaged endothelial sites. Since EPCs from patients with vascular disorders are impaired and inefficient, allogenic sources from adult or cord blood are considered as good alternatives. However, due to the reaction of immune system against allogenic cells which usually lead to their elimination, we focused on the exact role of EPCs on immune cells, particularly, T cells which are the most important cells applied in immune rejection. TNFα is one of the main activators of EPCs that recognizes two distinct receptors. TNFR1 is expressed ubiquitously and its interaction with TNFα leads to differentiation and apoptosis, whereas, TNFR2 is expressed predominantly on ECs, immune cells and neural cells and is involved in cell survival and proliferation. Interestingly, it has been shown that different immunosuppressive cells express TNFR2 and this is directly related to their immunosuppressive efficiency. However, little is known about immunological profile and function of TNFR2 in EPCs. METHODS: Using different in-vitro combinations, we performed co-cultures of ECs and T cells to investigate the immunological effect of EPCs on T cells. We interrupted in the TNFα/TNFR2 axis either by blocking the receptor using TNFR2 antagonist or blocking the ligand using T cells derived from TNFα KO mice. RESULTS: We demonstrated that EPCs are able to suppress T cell proliferation and modulate them towards less pro-inflammatory and active phenotypes. Moreover, we showed that TNFα/TNFR2 immune-checkpoint pathway is critical in EPC immunomodulatory effect. CONCLUSIONS: Our results reveal for the first time a mechanism that EPCs use to suppress immune cells, therefore, enabling them to form new immunosuppressive vessels. Furthermore, we have shown the importance of TNFα/TNFR2 axis in EPCs as an immune checkpoint pathway. We believe that targeting TNFR2 is especially crucial in cancer immune therapy since it controls two crucial aspects of tumor microenvironment: 1) Immunosuppression and 2) Angiogenesis. Video Abstract. (MP4 46355 kb).


Assuntos
Células Progenitoras Endoteliais , Terapia de Imunossupressão , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Linfócitos T/citologia , Fator de Necrose Tumoral alfa/imunologia , Adolescente , Adulto , Idoso , Animais , Células Cultivadas , Técnicas de Cocultura , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/imunologia , Feminino , Voluntários Saudáveis , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Transdução de Sinais , Adulto Jovem
13.
Basic Res Cardiol ; 115(2): 18, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980946

RESUMO

Circulating sca1+/flk1+ cells are hypothesized to be endothelial progenitor cells (EPCs) in mice that contribute to atheroprotection by replacing dysfunctional endothelial cells. Decreased numbers of circulating sca1+/flk1+ cells correlate with increased atherosclerotic lesions and impaired reendothelialization upon electric injury of the common carotid artery. However, legitimate doubts remain about the identity of the putative EPCs and their contribution to endothelial restoration. Hence, our study aimed to establish a phenotype for sca1+/flk1+ cells to gain a better understanding of their role in atherosclerotic disease. In wild-type mice, sca1+/flk1+ cells were mobilized into the peripheral circulation by granulocyte-colony stimulating factor (G-CSF) treatment and this movement correlated with improved endothelial regeneration upon carotid artery injury. Multicolor flow cytometry analysis revealed that sca1+/flk1+ cells predominantly co-expressed surface markers of conventional B cells (B2 cells). In RAG2-deficient mice and upon B2 cell depletion, sca1+/flk1+ cells were fully depleted. In the absence of monocytes, sca1+/flk1+ cell levels were unchanged. A PCR array focused on cell surface markers and next-generation sequencing (NGS) of purified sca1+/flk1+ cells confirmed their phenotype to be predominantly that of B cells. Finally, the depletion of B2 cells, including sca1+/flk1+ cells, in G-CSF-treated wild-type mice partly abolished the endothelial regenerating effect of G-CSF, indicating an atheroprotective role for sca1+/flk1+ B2 cells. In summary, we characterized sca1+/flk1+ cells as a subset of predominantly B2 cells, which are apparently involved in endothelial regeneration.


Assuntos
Antígenos Ly/metabolismo , Aterosclerose/metabolismo , Subpopulações de Linfócitos B/metabolismo , Lesões das Artérias Carótidas/metabolismo , Artéria Carótida Primitiva/metabolismo , Proliferação de Células , Células Progenitoras Endoteliais/metabolismo , Proteínas de Membrana/metabolismo , Reepitelização , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Antígenos Ly/genética , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/imunologia , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/imunologia , Artéria Carótida Primitiva/patologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Células Progenitoras Endoteliais/imunologia , Células Progenitoras Endoteliais/patologia , Feminino , Depleção Linfocítica , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
14.
Cardiovasc Revasc Med ; 21(2): 213-221, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31147259

RESUMO

BACKGROUND: Coronary stent neoatherosclerosis, thrombosis, and restenosis remain significant concerns with new-generation drug-eluting stents (DES). The Dual-Therapy CD34 antibody-covered sirolimus-eluting stent [dual therapy stent (DTS)] is a sirolimus-eluting stent with CD34 antibodies immobilized on its luminal surface to capture circulating endothelial progenitor cells and promote early endothelialization. We conducted a meta-analysis to determine whether the DTS was superior to standard DES. METHODS: We conducted a comprehensive search for controlled randomized and non-randomized studies. We presented data using risk ratios (95% confidence intervals) and measured heterogeneity using Higgins' I2. RESULTS: Five studies with a low risk of bias met the inclusion criteria, with a total of 1884 patients in the DTS and 1819 in standard DES arms. There was no difference between the 2 arms in the following 1-year outcomes: cardiac death [1% vs 0.9% RR 1.13 (95% CI 0.49-2.62) I2 = 0%], target lesion failure [6.2% vs 5.3% RR 1.12 (0.80-1.58) I2 = 0%], target lesion revascularization (TLR) [4.9% vs 3.4% RR 1.40 (0.93-2.10) I2 = 15%], target vessel failure [8.2% vs 6.1% RR 1.24 (0.75-2.04) I2 = 0%], target vessel myocardial infarction [1.1% vs 1.8% RR 0.73 (0.19-2.90) I2 = 62%] and stent thrombosis [0.4% vs 0.6% HR 0.85 (0.27-2.62) I2 = 0%]. However, compared with second-generation DES (EES and ZES), the DTS had significantly higher one-year TLR [5% vs. 3.1% RR 1.58 (1.02-2.46) P = 0.04 I2 = 0%]. CONCLUSION: One-year TLR was significantly higher in the DTS arm compared with second-generation DES. There was no difference in the other 1-year clinical outcomes compared with standard DES.


Assuntos
Anticorpos/administração & dosagem , Antígenos CD34/imunologia , Fármacos Cardiovasculares/administração & dosagem , Doença da Artéria Coronariana/terapia , Vasos Coronários/imunologia , Stents Farmacológicos , Células Progenitoras Endoteliais/imunologia , Intervenção Coronária Percutânea/instrumentação , Sirolimo/administração & dosagem , Anticorpos/efeitos adversos , Fármacos Cardiovasculares/efeitos adversos , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/imunologia , Trombose Coronária/etiologia , Vasos Coronários/diagnóstico por imagem , Humanos , Infarto do Miocárdio/etiologia , Intervenção Coronária Percutânea/efeitos adversos , Desenho de Prótese , Ensaios Clínicos Controlados Aleatórios como Assunto , Reepitelização , Fatores de Risco , Sirolimo/efeitos adversos , Fatores de Tempo , Resultado do Tratamento
15.
Sci Rep ; 9(1): 16752, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31727941

RESUMO

Moyamoya disease (MMD) is well known to be caused by insufficient cerebral vascular formation. However, the essential pathogenesis has not yet been identified. Using our recently developed technique of generating vasculogenic and anti-inflammatory cultures, we investigated endothelial progenitor cell (EPC) expansion and differentiation under the cytokine milieu generated by the peripheral blood mononuclear cells (PBMNCs) of the operated and non-operated MMD patients. EPC colony forming assay of the cultured PBMNCs disclosed the decline of the definitive EPC colony numbers in the both MMD patients. The level of interleukin-10 (IL-10) was lower in secretory cytokines from the cultured PBMNCs of MMD patients than that in that of controls using a cytometric bead array. The addition of human recombinant IL-10 to PBMNCs cultured from MMD patients restored the EPC colony forming potential of MMD PBMNCs. Following phorbol myristate acetate stimulation of the cultured PBMNCs, flow cytometry revealed a decrease in intracellular IL-10 storage in the main cell populations of the PBMNCs cultured from MMD patients relative to those cultured from controls. The present data provide the expected mechanism of vascular malformation in MMD pathogenesis originated from the insufficient production of IL-10 secreting cells from PBMNCs fostering EPC expansion and differentiation.


Assuntos
Células Progenitoras Endoteliais/citologia , Interleucina-10/metabolismo , Leucócitos Mononucleares/citologia , Macrófagos/citologia , Doença de Moyamoya/imunologia , Adulto , Estudos de Casos e Controles , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Regulação para Baixo , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/imunologia , Feminino , Citometria de Fluxo , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade , Doença de Moyamoya/patologia , Doença de Moyamoya/cirurgia , Acetato de Tetradecanoilforbol/farmacologia
16.
Int J Dermatol ; 58(12): 1398-1405, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31290139

RESUMO

BACKGROUND: One suggested reason for aberrant wound healing in keloid scars is chronic inflammation of the dermis. We hypothesized that excessive blood vessel formation and high capillary density in keloid tissue is caused by dysfunction of endothelial progenitor cells. METHODS: We compared the number of circulating endothelial progenitor cells and vasculogenic and angiogenic capacity, as well as secretory function, of circulating CD34+ cells in keloid patients and healthy individuals. RESULTS: Compared to mononuclear cell cultures from healthy donors, cultures of peripheral blood mononuclear cells obtained from keloid patients showed a more than twofold increase in the number of peripheral blood EPCs (fibronectin-adhering cells that phagocytized acetylated low-density lipoprotein and bound Ulex europaeus agglutinin-I lectin). However, there was no difference in colony-forming ability and participation in in vitro angiogenesis between circulating CD34+ cells isolated from keloid patients and healthy individuals. This means that circulating CD34+ /endothelial progenitor cells in keloid patients have normal vasculogenic and angiogenic function. However, CD34+ cells derived from keloid patients demonstrated a more than sevenfold expression of the interleukin-8 gene and a more than fivefold expression of the vascular endothelial growth factor gene than CD34+ cells derived from healthy individuals. CONCLUSIONS: These results support the role of vascular endothelial growth factor and interleukin-8 in increased recruitment of endothelial progenitor cells in keloid patients.


Assuntos
Células Progenitoras Endoteliais/imunologia , Interleucina-8/metabolismo , Queloide/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Idoso , Antígenos CD34/metabolismo , Contagem de Células , Diferenciação Celular , Células Cultivadas , Células Progenitoras Endoteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Voluntários Saudáveis , Humanos , Queloide/sangue , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , Cicatrização/imunologia , Adulto Jovem
17.
Circ J ; 83(7): 1422-1430, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31178469

RESUMO

In 1997, the seminal manuscript by Asahara, Murohara, Isner et al outlined the evidence for the existence of circulating, bone marrow-derived cells capable of stimulating and contributing to the formation of new blood vessels. Consistent with the paradigm shift that this work represented, it triggered much scientific debate and controversy, some of which persists 2 decades later. In contrast, the clinical application of autologous CD34 cell therapy has been marked by a track record of consistent safety and clinical benefit in multiple ischemic conditions. In this review, we summarize the preclinical and clinical evidence from over 700 patients in clinical trials of CD34 cell therapy.


Assuntos
Antígenos CD34/imunologia , Transtornos Cerebrovasculares/cirurgia , Células Progenitoras Endoteliais/transplante , Extremidade Inferior/irrigação sanguínea , Isquemia Miocárdica/cirurgia , Neovascularização Fisiológica , Doença Arterial Periférica/cirurgia , Regeneração , Transplante de Células-Tronco , Animais , Transtornos Cerebrovasculares/diagnóstico , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/fisiopatologia , Células Progenitoras Endoteliais/imunologia , Humanos , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/imunologia , Isquemia Miocárdica/fisiopatologia , Doença Arterial Periférica/diagnóstico , Doença Arterial Periférica/imunologia , Doença Arterial Periférica/fisiopatologia , Recuperação de Função Fisiológica , Transplante de Células-Tronco/efeitos adversos , Resultado do Tratamento
18.
Am J Med Sci ; 357(1): 49-56, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30611320

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) are special types of stem cells and are a potential novel therapeutic approach in acute lung injury (ALI). Transplantation of EPCs can ameliorate the inflammatory state by reducing adhesion and exudation of inflammatory cells. However, the mechanism underlying the effect of EPCs on inflammatory response modulation remains unclear. The aim of the present study was to investigate the effect of EPCs on the modulation of neutrophils in vitro and in vivo. MATERIALS AND METHODS: EPCs were cocultured with neutrophils after lipopolysaccharide stimulation in vitro or transplanted into ALI rats, and neutrophil inflammatory mediators including tumor necrosis factor-α, interleukin-1ß, neutrophil elastase, myeloperoxidase and matrix metalloproteinases-9 were detected by enzyme-linked immunosorbent assay, an myeloperoxidase detection kits, reverse transcription-polymerase chain reaction and western blotting. RESULTS: The results showed that EPCs significantly downregulated the expression of inflammatory mediators when cocultured with neutrophils in vitro or in vivo. CONCLUSIONS: These findings demonstrated that EPCs contributed to lung injury in ALI rats by downregulating neutrophil inflammatory mediators.


Assuntos
Lesão Pulmonar Aguda/imunologia , Células Progenitoras Endoteliais/transplante , Inflamação/prevenção & controle , Neutrófilos/imunologia , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Células Progenitoras Endoteliais/imunologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Neutrófilos/metabolismo , Ratos , Ratos Sprague-Dawley
19.
Cardiol J ; 26(3): 283-291, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29611172

RESUMO

BACKGROUND: Even with drug-eluting stents, the risk of in-stent restenosis (ISR) remains high. The goal of this study was to investigate the use of an endothelial progenitor cell (EPC) capture stent plus regional EPC transplantation to reduce the ISR rate. METHODS: Endothelial progenitor cell capture stents were fabricated using fibrin gel and anti-CD34 plus anti-VEGFR-2 dual antibodies. Twenty male New Zealand white rabbits established as an atherosclerotic model were randomly divided into two groups: group 1 (n = 10), in which EPC capture stents were deployed into the right iliac artery; and group 2 (n = 10), in which sirolimus-eluting stents were placed. In both groups, EPCs were transplanted into target vessels beyond the stents, with outflow blocked. Radiologic-pathologic correlation outcomes were reviewed after 2 months. RESULTS: The technical success rate of EPC capture stent placement plus EPC transplantation was 100%. The ISR rate in group 1 was lower than in group 2 (1/10 vs. 4/10; p > 0.05). Minimal luminal diameters were larger in group 1 than in group 2 (computed tomographic angiography, 1.85 ± 0.15 mm vs. 1.50 ± 0.20 mm; duplex ultrasound, 1.90 ± 0.10 mm vs. 1.70 ± 0.30 mm; p > 0.05). Transplanted EPCs were tracked positively only in group 1. Pathologic analysis demonstrated neointimal hyperplasia thickness of 0.21 ± 0.09 mm in group 1 vs. 0.11 ± 0.07 mm in group 2 (p < 0.05). CONCLUSION: Endothelial progenitor cell capture stent placement plus local EPC transplant decreases the ISR rate through thrombosis reduction rather than through neointimal hyperplasia inhibition.


Assuntos
Angioplastia com Balão/instrumentação , Aterosclerose/terapia , Materiais Revestidos Biocompatíveis , Células Progenitoras Endoteliais/transplante , Artéria Ilíaca/patologia , Placa Aterosclerótica , Stents , Animais , Anticorpos/metabolismo , Antígenos CD34/imunologia , Antígenos CD34/metabolismo , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Células Cultivadas , Modelos Animais de Doenças , Células Progenitoras Endoteliais/imunologia , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Fibronectinas/metabolismo , Artéria Ilíaca/imunologia , Artéria Ilíaca/metabolismo , Masculino , Desenho de Prótese , Coelhos , Recidiva , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
20.
Cardiovasc Revasc Med ; 20(9): 739-743, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30442536

RESUMO

BACKGROUND/PURPOSE: Primary percutaneous coronary intervention (PCI) during acute ST-segment elevation myocardial infarction (STEMI) represents a thrombotic milieu and is associated with delayed healing after stenting. The pro-healing combination sirolimus eluting endothelial progenitor cell (EPC) capture stents encourage early endothelialization after stenting and may be beneficial in the STEMI population. We aim to evaluate the clinical outcomes one year and beyond for patients with STEMI who received the combination sirolimus eluting EPC capture stents during primary PCI. METHODS/MATERIAL: All STEMI patients implanted with combination sirolimus eluting EPC capture stents during primary PCI from November 2013 to December 2016 were enrolled. The primary outcome was target lesion failure (TLF) at in-hospital, one-month, one-year and beyond one year. RESULTS: A total of 260 consecutive STEMI patients (283 lesions) were implanted with 313 combination sirolimus eluting EPC capture stents during primary PCI. Mean age was 56.1 ±â€¯11.2 years and 88.8% were male. One in ten patients (10.9%) had cardiogenic shock on presentation, 7.3% needed mechanical ventilation and 7.7% had intra-aortic balloon pump inserted. A total of 97.9% of lesions achieve final TIMI 3 flow. Device success was seen in all patients. At extended follow up period (median 23.4 months), the clinical outcomes were TLF 8.8%, major adverse cardiovascular events 10.8%, cardiac mortality 4.2%, target vessel myocardial infarction 3.4%, target lesion revascularization 3.8%, and definite stent thrombosis 1.9%. CONCLUSIONS: This study demonstrated acceptable clinical outcomes for an all-comers STEMI patients undergoing primary PCI with the use of combination sirolimus eluting EPC cell capture stents.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antígenos CD34/imunologia , Fármacos Cardiovasculares/administração & dosagem , Doença da Artéria Coronariana/terapia , Stents Farmacológicos , Células Progenitoras Endoteliais/imunologia , Intervenção Coronária Percutânea/instrumentação , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Sirolimo/administração & dosagem , Idoso , Anticorpos Monoclonais/efeitos adversos , Fármacos Cardiovasculares/efeitos adversos , Adesão Celular , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/imunologia , Doença da Artéria Coronariana/mortalidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Intervenção Coronária Percutânea/efeitos adversos , Desenho de Prótese , Sistema de Registros , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico por imagem , Infarto do Miocárdio com Supradesnível do Segmento ST/imunologia , Infarto do Miocárdio com Supradesnível do Segmento ST/mortalidade , Sirolimo/efeitos adversos , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA