Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38732080

RESUMEN

Endothelial progenitor cells (EPCs) play a critical role in cardiovascular regeneration. Enhancement of their native properties would be highly beneficial to ensuring the proper functioning of the cardiovascular system. As androgens have a positive effect on the cardiovascular system, we hypothesized that dihydrotestosterone (DHT) could also influence EPC-mediated repair processes. To evaluate this hypothesis, we investigated the effects of DHT on cultured human EPCs' proliferation, viability, morphology, migration, angiogenesis, gene and protein expression, and ability to integrate into cardiac tissue. The results showed that DHT at different concentrations had no cytotoxic effect on EPCs, significantly enhanced the cell proliferation and viability and induces fast, androgen-receptor-dependent formation of capillary-like structures. DHT treatment of EPCs regulated gene expression of androgen receptors and the genes and proteins involved in cell migration and angiogenesis. Importantly, DHT stimulation promoted EPC migration and the cells' ability to adhere and integrate into murine cardiac slices, suggesting it has a role in promoting tissue regeneration. Mass spectrometry analysis further highlighted the impact of DHT on EPCs' functioning. In conclusion, DHT increases the proliferation, migration, and androgen-receptor-dependent angiogenesis of EPCs; enhances the cells' secretion of key factors involved in angiogenesis; and significantly potentiates cellular integration into heart tissue. The data offer support for potential therapeutic applications of DHT in cardiovascular regeneration and repair processes.


Asunto(s)
Movimiento Celular , Dihidrotestosterona , Células Progenitoras Endoteliales , Sangre Fetal , Receptores Androgénicos , Sangre Fetal/citología , Dihidrotestosterona/farmacología , Humanos , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Proliferación Celular , Supervivencia Celular , Expresión Génica , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas de la Membrana/genética , Metaloproteinasa 9 de la Matriz/genética , Basigina/genética , Animales , Ratones , Ventrículos Cardíacos/citología , Movimiento Celular/efectos de los fármacos
2.
Int J Mol Sci ; 25(10)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38791147

RESUMEN

Despite neutrophil involvement in inflammation and tissue repair, little is understood about their inflammatory status in acute coronary syndrome (ACS) patients with poor outcomes. Hence, we investigated the potential correlation between neutrophil inflammatory markers and the prognosis of ACS patients with/without diabetes and explored whether neutrophils demonstrate a unique inflammatory phenotype in patients experiencing an adverse in-hospital outcome. The study enrolled 229 ACS patients with or without diabetes. Poor evolution was defined as either death, left ventricular ejection fraction (LVEF) <40%, Killip Class 3/4, ventricular arrhythmias, or mechanical complications. Univariate and multivariate analyses were employed to identify clinical and paraclinical factors associated with in-hospital outcomes. Neutrophils isolated from fresh blood were investigated using qPCR, Western blot, enzymatic assay, and immunofluorescence. Poor evolution post-myocardial infarction (MI) was associated with increased number, activity, and inflammatory status of neutrophils, as indicated by significant increase of Erythrocyte Sedimentation Rate (ESR), C-reactive protein (CRP), fibrinogen, interleukin-1ß (IL-1ß), and, interleukin-6 (IL-6). Among the patients with complicated evolution, neutrophil activity had an important prognosis value for diabetics. Neutrophils from patients with unfavorable evolution revealed a pro-inflammatory phenotype with increased expression of CCL3, IL-1ß, interleukin-18 (IL-18), S100A9, intracellular cell adhesion molecule-1 (ICAM-1), matrix metalloprotease (MMP-9), of molecules essential in reactive oxygen species (ROS) production p22phox and Nox2, and increased capacity to form neutrophil extracellular traps. Inflammation is associated with adverse short-term prognosis in acute ACS, and inflammatory biomarkers exhibit greater specificity in predicting short-term outcomes in diabetics. Moreover, neutrophils from patients with unfavorable evolution exhibit distinct inflammatory patterns, suggesting that alterations in the innate immune response in this subgroup may exert detrimental effects on disease progression.


Asunto(s)
Síndrome Coronario Agudo , Inflamación , Neutrófilos , Humanos , Neutrófilos/metabolismo , Neutrófilos/inmunología , Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/complicaciones , Masculino , Femenino , Pronóstico , Persona de Mediana Edad , Anciano , Inflamación/sangre , Inflamación/patología , Biomarcadores/sangre , Diabetes Mellitus/sangre , Diabetes Mellitus/inmunología , Diabetes Mellitus/patología
3.
Int J Mol Sci ; 23(16)2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-36012192

RESUMEN

Adipose-derived mesenchymal stromal cells (ADSC) are a promising source for cellular therapy of chronic wounds. However, the limited life span during in vitro expansion impedes their extensive use in clinical applications and basic research. We hypothesize that by introduction of an ectopic expression of telomerase into ADSC, the cells' lifespans could be significantly extended. To test this hypothesis, we aimed at engineering an immortalized human ADSC line using a lentiviral transduction with human telomerase (hTERT). ADSC were transduced with a third-generation lentiviral system and a hTERT codifying plasmid (pLV-hTERT-IRES-hygro). A population characterized by increased hTERT expression, extensive proliferative potential and remarkable (potent) multilineage differentiation capacity was selected. The properties for wound healing of this immortalized ADSC line were assessed after 17 passages. Their secretome induced the proliferation and migration of keratinocytes, dermal fibroblasts, and endothelial cells similarly to untransduced ADSC. Moreover, they sustained the complete re-epithelialization of a full thickness wound performed on a skin organotypic model. In summary, the engineered immortalized ADSC maintain the beneficial properties of parent cells and could represent a valuable and suitable tool for wound healing in particular, and for skin regenerative therapy in general.


Asunto(s)
Células Madre Mesenquimatosas , Telomerasa , Proliferación Celular , Células Endoteliales/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Cicatrización de Heridas/fisiología
4.
Int J Mol Sci ; 23(9)2022 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-35563680

RESUMEN

Prognosis after myocardial infarction (MI) varies greatly depending on the extent of damaged area and the management of biological processes during recovery. Reportedly, the inhibition of the pro-inflammatory S100A9 reduces myocardial damage after MI. We hypothesize that a S100A9 blockade induces changes of major signaling pathways implicated in post-MI healing. Mass spectrometry-based proteomics and gene analyses of infarcted mice left ventricle were performed. The S100A9 blocker (ABR-23890) was given for 3 days after coronary ligation. At 3 and 7 days post-MI, ventricle samples were analyzed versus control and Sham-operated mice. Blockade of S100A9 modulated the expressed proteins involved in five biological processes: leukocyte cell-cell adhesion, regulation of the muscle cell apoptotic process, regulation of the intrinsic apoptotic signaling pathway, sarcomere organization and cardiac muscle hypertrophy. The blocker induced regulation of 36 proteins interacting with or targeted by the cellular tumor antigen p53, prevented myocardial compensatory hypertrophy, and reduced cardiac markers of post-ischemic stress. The blockade effect was prominent at day 7 post-MI when the quantitative features of the ventricle proteome were closer to controls. Blockade of S100A9 restores key biological processes altered post-MI. These processes could be valuable new pharmacological targets for the treatment of ischemic heart. Mass spectrometry data are available via ProteomeXchange with identifier PXD033683.


Asunto(s)
Infarto del Miocardio , Proteoma , Alarminas/metabolismo , Animales , Calgranulina B/genética , Calgranulina B/metabolismo , Ventrículos Cardíacos/metabolismo , Hipertrofia/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Proteoma/metabolismo , Transducción de Señal , Remodelación Ventricular
5.
Int J Mol Sci ; 23(7)2022 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-35409184

RESUMEN

Calcific aortic valve disease (CAVD) is a progressive inflammatory disorder characterized by extracellular matrix remodeling and valvular interstitial cells (VIC) osteodifferentiation leading to valve leaflets calcification and impairment movement. Runx2, the master transcription factor involved in VIC osteodifferentiation, modulates the expression of other osteogenic molecules. Previously, we have demonstrated that the osteoblastic phenotypic shift of cultured VIC is impeded by Runx2 silencing using fullerene (C60)-polyethyleneimine (PEI)/short hairpin (sh)RNA-Runx2 (shRunx2) polyplexes. Since the use of polyplexes for in vivo delivery is limited by their instability in the plasma and the non-specific tissue interactions, we designed and obtained targeted, lipid-enveloped polyplexes (lipopolyplexes) suitable for (1) systemic administration and (2) targeted delivery of shRunx2 to osteoblast-differentiated VIC (oVIC). Vascular cell adhesion molecule (VCAM)-1 expressed on the surface of oVIC was used as a target, and a peptide with high affinity for VCAM-1 was coupled to the surface of lipopolyplexes encapsulating C60-PEI/shRunx2 (V-LPP/shRunx2). We report here that V-LPP/shRunx2 lipopolyplexes are cyto- and hemo-compatible and specifically taken up by oVIC. These lipopolyplexes are functional as they downregulate the Runx2 gene and protein expression, and their uptake leads to a significant decrease in the expression of osteogenic molecules (OSP, BSP, BMP-2). These results identify V-LPP/shRunx2 as a new, appropriately directed vehicle that could be instrumental in developing novel strategies for blocking the progression of CAVD using a targeted nanomedicine approach.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/metabolismo , Calcinosis/metabolismo , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Osteoblastos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
6.
Int J Mol Sci ; 23(7)2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35409134

RESUMEN

Parathyroid hormone (PTH) is a key regulator of calcium, phosphate and vitamin D metabolism. Although it has been reported that aortic valve calcification was positively associated with PTH, the pathophysiological mechanisms and the direct effects of PTH on human valvular cells remain unclear. Here we investigated if PTH induces human valvular endothelial cells (VEC) dysfunction that in turn could impact the switch of valvular interstitial cells (VIC) to an osteoblastic phenotype. Human VEC exposed to PTH were analyzed by qPCR, western blot, Seahorse, ELISA and immunofluorescence. Our results showed that exposure of VEC to PTH affects VEC metabolism and functions, modifications that were accompanied by the activation of p38MAPK and ERK1/2 signaling pathways and by an increased expression of osteogenic molecules (BMP-2, BSP, osteocalcin and Runx2). The impact of dysfunctional VEC on VIC was investigated by exposure of VIC to VEC secretome, and the results showed that VIC upregulate molecules associated with osteogenesis (BMP-2/4, osteocalcin and TGF-ß1) and downregulate collagen I and III. In summary, our data show that PTH induces VEC dysfunction, which further stimulates VIC to differentiate into a pro-osteogenic pathological phenotype related to the calcification process. These findings shed light on the mechanisms by which PTH participates in valve calcification pathology and suggests that PTH and the treatment of hyperparathyroidism represent a therapeutic strategy to reduce valvular calcification.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/patología , Calcinosis/patología , Diferenciación Celular/genética , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogénesis , Hormona Paratiroidea/metabolismo , Fenotipo
7.
J Cell Mol Med ; 25(20): 9483-9495, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34561944

RESUMEN

Valve disease and particularly calcific aortic valve disease (CAVD) and diabetes (DM) are progressive diseases constituting a global health burden for all aging societies (Progress in Cardiovascular Diseases. 2014;56(6):565: Circulation Research. 2021;128(9):1344). Compared to non-diabetic individuals (The Lancet. 2008;371(9626):1800: The American Journal of Cardiology. 1983;51(3):403: Journal of the American College of Cardiology. 2017;69(12):1523), the diabetic patients have a significantly greater propensity for cardiovascular disorders and faster degeneration of implanted bioprosthetic aortic valves. Previously, using an original experimental model, the diabetic-hyperlipemic hamsters, we have shown that the earliest alterations induced by these conditions occur at the level of the aortic valves and, with time these changes lead to calcifications and CAVD. However, there are no pharmacological treatments available to reverse or retard the progression of aortic valve disease in diabetes, despite the significant advances in the field. Therefore, it is critical to uncover the mechanisms of valve disease progression, find biomarkers for diagnosis and new targets for therapies. This review aims at presenting an update on the basic research in CAVD in the context of diabetes. We provide an insight into the accumulated data including our results on diabetes-induced progressive cell and molecular alterations in the aortic valve, new potential biomarkers to assess the evolution and therapy of the disease, advancement in targeted nanotherapies, tissue engineering and the potential use of circulating endothelial progenitor cells in CAVD.


Asunto(s)
Válvula Aórtica/patología , Complicaciones de la Diabetes , Diabetes Mellitus/metabolismo , Enfermedades de las Válvulas Cardíacas/etiología , Enfermedades de las Válvulas Cardíacas/metabolismo , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/ultraestructura , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores , Terapia Combinada , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales/ultraestructura , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Enfermedades de las Válvulas Cardíacas/diagnóstico , Enfermedades de las Válvulas Cardíacas/terapia , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Mediadores de Inflamación/metabolismo
8.
Int J Mol Sci ; 22(11)2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-34073119

RESUMEN

Cells convey information among one another. One instrument employed to transmit data and constituents to specific (target) cells is extracellular vesicles (EVs). They originate from a variety of cells (endothelial, immune cells, platelets, mesenchymal stromal cells, etc.), and consequently, their surface characteristics and cargo vary according to the paternal cell. The cargo could be DNA, mRNA, microRNA, receptors, metabolites, cytoplasmic proteins, or pathological molecules, as a function of which EVs exert different effects upon endocytosis in recipient cells. Recently, EVs have become important participants in a variety of pathologies, including atherogenesis and coronavirus disease 2019 (COVID-19)-associated thrombosis. Herein, we summarize recent advances and some of our own results on the role of EVs in atherosclerotic cardiovascular diseases, and discuss their potential to function as signaling mediators, biomarkers and therapeutic agents. Since COVID-19 patients have a high rate of thrombotic events, a special section of the review is dedicated to the mechanism of thrombosis and the possible therapeutic potential of EVs in COVID-19-related thrombosis. Yet, EV mechanisms and their role in the transfer of information between cells in normal and pathological conditions remain to be explored.


Asunto(s)
Aterosclerosis/metabolismo , COVID-19/metabolismo , Vesículas Extracelulares/metabolismo , Trombosis/metabolismo , Aterosclerosis/fisiopatología , Aterosclerosis/terapia , Aterosclerosis/virología , Biomarcadores/metabolismo , COVID-19/complicaciones , COVID-19/fisiopatología , COVID-19/terapia , Células Endoteliales/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/virología , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal/inmunología , Trombosis/complicaciones , Trombosis/fisiopatología , Trombosis/virología
9.
Int J Mol Sci ; 22(13)2021 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-34281252

RESUMEN

Cardiovascular diseases have attracted our full attention not only because they are the main cause of mortality and morbidity in many countries but also because the therapy for and cure of these maladies are among the major challenges of the medicine in the 21st century [...].


Asunto(s)
Enfermedades Cardiovasculares/etiología , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Vesículas Extracelulares/metabolismo , Marcadores Genéticos , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Modelos Cardiovasculares , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Factores de Riesgo
10.
Int J Mol Sci ; 22(12)2021 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-34207905

RESUMEN

Bone marrow-derived mesenchymal stromal cells (MSCs) are major players in regenerative therapies for wound healing via their paracrine activity, mediated partially by exosomes. Our purpose was to test if MSC-derived exosomes could accelerate wound healing by enhancing the biological properties of the main cell types involved in the key phases of this process. Thus, the effects of exosomes on (i) macrophage activation, (ii) angiogenesis, (iii) keratinocytes and dermal fibroblasts proliferation and migration, and (iv) the capacity of myofibroblasts to regulate the turnover of the extracellular matrix were evaluated. The results showed that, although exosomes did not exhibit anti-inflammatory properties, they stimulated angiogenesis. Exposure of keratinocytes and dermal (myo)fibroblasts to exosomes enhanced their proliferation and migratory capacity. Additionally, exosomes prevented the upregulation of gene expression for type I and III collagen, α-smooth muscle actin, and MMP2 and 14, and they increased MMP13 expression during the fibroblast-myofibroblast transition. The regenerative properties of exosomes were validated using a wound healing skin organotypic model, which exhibited full re-epithelialization upon exosomes exposure. In summary, these data indicate that exosomes enhance the biological properties of keratinocytes, fibroblasts, and endothelial cells, thus providing a reliable therapeutic tool for skin regeneration.


Asunto(s)
Exosomas/metabolismo , Fibroblastos/metabolismo , Queratinocitos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Piel/metabolismo , Cicatrización de Heridas , Humanos , Piel/lesiones
11.
Int J Mol Sci ; 22(11)2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34073327

RESUMEN

Mesenchymal stromal cells (MSC) are promising candidates for regenerative therapy of the infarcted heart. However, poor cell retention within the transplantation site limits their potential. We hypothesized that MSC benefits could be enhanced through a dual-cell approach using jointly endothelial colony forming cells (ECFC) and MSC. To assess this, we comparatively evaluated the effects of the therapy with MSC and ECFC versus MSC-only in a mouse model of myocardial infarction. Heart function was assessed by echocardiography, and the molecular crosstalk between MSC and ECFC was evaluated in vitro through direct or indirect co-culture systems. We found that dual-cell therapy improved cardiac function in terms of ejection fraction and stroke volume. In vitro experiments showed that ECFC augmented MSC effector properties by increasing Connexin 43 and Integrin alpha-5 and the secretion of healing-associated molecules. Moreover, MSC prompted the organization of ECFC into vascular networks. This indicated a reciprocal modulation in the functionality of MSC and ECFC. In conclusion, the crosstalk between MSC and ECFC augments the therapeutic properties of MSC and enhances the angiogenic properties of ECFC. Our data consolidate the dual-cell therapy as a step forward for the development of effective treatments for patients affected by myocardial infarction.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Infarto del Miocardio , Miocardio , Volumen Sistólico , Animales , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Femenino , Xenoinjertos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Miocardio/patología
12.
J Cell Mol Med ; 24(20): 12131-12140, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32935914

RESUMEN

Nephropathy is a major chronic complication of diabetes. A crucial role in renal pathophysiology is played by hydrogen sulphide (H2 S) that is produced excessively by the kidney; however, the data regarding H2 S bioavailability are inconsistent. We hypothesize that early type 1 diabetes (T1D) increases H2 S production by a mechanism involving hyperglycaemia-induced alterations in sulphur metabolism. Plasma and kidney tissue collected from T1D double transgenic mice were subjected to mass spectrometry-based proteomic analysis, and the results were validated by immunological and gene expression assays.T1D mice exhibited a high concentration of H2 S in the plasma and kidney tissue and histological, showed signs of subtle kidney fibrosis, characteristic for early renal disease. The shotgun proteomic analyses disclosed that the level of enzymes implicated in sulphate activation modulators, H2 S-oxidation and H2 S-production were significantly affected (ie 6 up-regulated and 4 down-regulated). Gene expression results corroborated well with the proteomic data. Dysregulation of H2 S enzymes underly the changes occurring in H2 S production, which in turn could play a key role in the initiation of renal disease. The new findings lead to a novel target in the therapy of diabetic nephropathy. Mass spectrometry data are available via ProteomeXchange with identifier PXD018053.


Asunto(s)
Nefropatías Diabéticas/enzimología , Riñón/metabolismo , Azufre/metabolismo , Animales , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/patología , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Sulfuro de Hidrógeno/metabolismo , Redes y Vías Metabólicas , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados
13.
J Cell Mol Med ; 24(17): 9692-9704, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32666712

RESUMEN

The development of stem cell technology in combination with advances in biomaterials has opened new ways of producing engineered tissue substitutes. In this study, we investigated whether the therapeutic potential of an acellular porous scaffold made of type I collagen can be improved by the addition of a powerful trophic agent in the form of mesenchymal stromal cells conditioned medium (MSC-CM) in order to be used as an acellular scaffold for skin wound healing treatment. Our experiments showed that MSC-CM sustained the adherence of keratinocytes and fibroblasts as well as the proliferation of keratinocytes. Moreover, MSC-CM had chemoattractant properties for keratinocytes and endothelial cells, attributable to the content of trophic and pro-angiogenic factors. Also, for the dermal fibroblasts cultured on collagen scaffold in the presence of MSC-CM versus serum control, the ratio between collagen III and I mRNAs increased by 2-fold. Furthermore, the gene expression for α-smooth muscle actin, tissue inhibitor of metalloproteinase-1 and 2 and matrix metalloproteinase-14 was significantly increased by approximately 2-fold. In conclusion, factors existing in MSC-CM improve the colonization of collagen 3D scaffolds, by sustaining the adherence and proliferation of keratinocytes and by inducing a pro-healing phenotype in fibroblasts.


Asunto(s)
Colágeno/metabolismo , Células HaCaT/metabolismo , Células Madre Mesenquimatosas/metabolismo , Piel/metabolismo , Técnicas de Cultivo de Célula/métodos , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Ingeniería de Tejidos/métodos , Andamios del Tejido , Cicatrización de Heridas/fisiología
14.
J Cell Mol Med ; 24(18): 10889-10897, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32785979

RESUMEN

Subcutaneous transplantation of mesenchymal stromal cells (MSC) emerged as an alternative to intravenous administration because it avoids the pulmonary embolism and prolongs post-transplantation lifetime. The goal of this study was to investigate the mechanisms by which these cells could affect remote organs. To this aim, murine bone marrow-derived MSC were subcutaneously transplanted in different anatomical regions and the survival and behaviour have been followed. The results showed that upon subcutaneous transplantation in mice, MSC formed multicellular aggregates and did not migrate significantly from the site of injection. Our data suggest an important role of hypoxia-inducible signalling pathways in stimulating local angiogenesis and the ensuing modulation of the kinetics of circulating cytokines with putative protective effects at distant sites. These data expand the current understanding of cell behaviour after subcutaneous transplantation and contribute to the development of a non-invasive cell-based therapy for distant organ protection.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Tejido Subcutáneo/fisiología , Tejido Adiposo Pardo , Tejido Adiposo Blanco , Animales , Agregación Celular , Hipoxia de la Célula , Células Cultivadas , Microambiente Celular , Citocinas/sangre , Supervivencia de Injerto , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Especificidad de Órganos , Organismos Libres de Patógenos Específicos , Grasa Subcutánea , Tejido Subcutáneo/irrigación sanguínea , Trasplante Heterotópico
15.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-32825521

RESUMEN

Fas ligand (First apoptosis signal ligand, FasL, also known as CD95L) is the common executioner of apoptosis within the tumor necrosis factor (TNF) superfamily. We aimed to induce functional FasL expression in transduced cells using an adenovirus vector, which has the advantage of strong and transient induction of the gene included in the adenoviral genome. Here, we report that the adenovirus carrying a truncated FasL gene, named FasL minigene, encoding the full-length FasL protein (Ad-gFasL) is more efficient than the adenovirus carrying FasL cDNA (Ad-cFasL) in the induction of FasL expression in transduced cells. FasL minigene (2887 bp) lacking the second intron and a part of the 3'-UTR was created to reduce the gene length due to the size limitation of the adenoviral genome. The results show that, in transduced hepatocytes, strong expression of mRNA FasL appeared after 10 h for Ad-gFasL, while for Ad-cFasL, a faint expression appeared after 16 h. For Ad-gFasL, the protein expression was noticed starting with 0.5 transfection units (TU)/cell, while for Ad-cFasL, it could not be revealed. FasL-expressing endothelial cells induced apoptosis of A20 cells in co-culture experiments. FasL-expressing cells may be exploitable in various autoimmune diseases such as graft-versus-host disease, chronic colitis, and type I diabetes.


Asunto(s)
Adenoviridae/genética , Proteína Ligando Fas/genética , Técnicas de Transferencia de Gen , Regiones no Traducidas 3' , Animales , Apoptosis , Bovinos , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/fisiología , Proteína Ligando Fas/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hepatocitos/fisiología , Humanos , Intrones , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transgenes
16.
Int J Mol Sci ; 21(24)2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-33339419

RESUMEN

There is an intensive effort to identify biomarkers to predict cardiovascular disease evolution. We aimed to determine the potential of microRNAs to predict the appearance of cardiovascular events (CVEs) in patients with peripheral artery disease (PAD) following femoral artery bypass surgery. Forty-seven PAD patients were enrolled and divided into two groups, without CVEs (n = 35) and with CVEs (n = 12), during 1 year follow-up. Intra-surgery atherosclerotic plaques from femoral arteries were collected and the levels of miR-142, miR-223, miR-155, and miR-92a of the primary transcripts of these microRNAs (pri-miRNAs), and gene expression of Drosha and Dicer were determined. Results showed that, in the plaques, miR-142, miR-223, and miR-155 expression levels were significantly increased in PAD patients with CVEs compared to those without CVEs. Positive correlations between these miRNAs and their pri-miRNAs levels and the Dicer/Drosha expression were observed. In the plasma of PAD patients with CVEs compared to those without CVEs, miR-223 and miR-142 were significantly increased. The multiple linear regression analyses revealed significant associations among several plasma lipids, oxidative and inflammatory parameters, and plasma miRNAs levels. Receiver operator characteristic (ROC) analysis disclosed that plasma miR-142 levels could be an independent predictor for CVEs in PAD patients. Functional bioinformatics analyses supported the role of these miRNAs in the regulation of biological processes associated with atherosclerosis. Taken together, these data suggest that plasma levels of miR-142, miR-223, miR-155, and miR-92a can significantly predict CVEs among PAD patients with good accuracy, and that plasma levels of miR-142 can be an independent biomarker to predict post-surgery CVEs development in PAD patients.


Asunto(s)
MicroARNs/sangre , Enfermedad Arterial Periférica/sangre , Placa Aterosclerótica/sangre , Complicaciones Posoperatorias/sangre , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Biomarcadores/metabolismo , Femenino , Arteria Femoral/cirugía , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/cirugía , Placa Aterosclerótica/metabolismo , Complicaciones Posoperatorias/metabolismo , Injerto Vascular/efectos adversos
17.
Int J Mol Sci ; 22(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396269

RESUMEN

Mesenchymal stromal cells (MSC) display several mechanisms of action that may be harnessed for therapeutic purposes. One of their most attractive features is their immunomodulatory activity that has been extensively characterized both in vitro and in vivo. While this activity has proven to be very efficient, it is transient. We aimed to enhance it by transforming MSC to overexpress a first apoptosis signal (Fas) ligand (FasL). In this study, our goal was to induce FasL overexpression through adenoviral transduction in MSC to improve their immunomodulatory activity. We characterized the impact of FasL overexpression on the morphology, proliferation, viability, phenotype, multilineage differentiation potential and immunomodulation of MSC. Moreover, we determined their suppressive properties in mixed reactions with A20 cells, as well as with stimulated splenocytes. Our findings demonstrate that FasL-overexpressing MSC exhibit improved immunosuppressive properties, while maintaining their MSC-characteristic features. In conclusion, we establish, in a proof-of-concept set-up, that FasL-overexpressing MSC represent good candidates for therapeutic intervention targeted at autoimmune disorders.


Asunto(s)
Apoptosis , Proteína Ligando Fas/metabolismo , Inmunomodulación , Células Madre Mesenquimatosas/inmunología , Bazo/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Proteína Ligando Fas/genética , Femenino , Activación de Linfocitos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD , Bazo/citología , Bazo/metabolismo
18.
J Mol Cell Cardiol ; 133: 115-124, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31201797

RESUMEN

Scarcity of gender specific donor hearts highlights the importance of mesenchymal stem cells (MSCs) as a therapeutic tool for heart repair. However, inefficient incorporation, retention, and activity of MSCs in cardiac tissue remain an obstacle. Since surges in follicular estradiol (E2; µmolar-range) trigger tissue remodeling (e.g. ovulation) and E2 exerts beneficial actions on the cardiovascular system, we hypothesized that E2 may promote/improve MSC-mediated cardiac repair processes. Using Wharton's jelly (WJ)-derived MSCs we assessed the effects of E2 on MSC proliferation, directed migration, and engraftment in murine heart slices (using xCELLigence real-time cell-impedance system, DNA quantification, and microscopy) and on MSC-induced angiogenesis in vivo (matrigel plug assay). Protein expression was assessed by Western blotting, ELISA/Luminex, and proteomic analysis; whereas mRNA expression was assessed by qRT-PCR. MSCs expressed estrogen receptors (ERs) -alpha and -beta. E2 promoted MSC proliferation and up-regulated mRNA and protein expression of ER-alpha, ER-beta, extracellular matrix metalloproteinase inducer (EMMPRIN), and matrix metalloproteinase (MMP) -9, yet down-regulated MMP-2 expression. Moreover, E2 up-regulated expression of vascular endothelial growth factor (VEGF)-A, VEGFR-2, vascular cell adhesion protein-1 (VCAM-1), and angiogenin (ANG) and stimulated nitric oxide (NO) production via ER. Proteomic analysis of MSCs showed that E2 up-regulated 47 proteins, down-regulated 7 proteins, and increased the expression of key biochemical components/pathways involved in tissue repair. In MSCs co-cultured with murine heart-slices, E2 significantly induced MSC migration in an ER-alpha-dependent fashion and significantly increased the secretion of MMP-2, MMP-9, ANG, and VEGF. In an in vivo matrigel assay, E2-treated MSCs increased angiogenesis and hemoglobin content. In conclusion, E2-treatment increases the incorporation of MSCs in heart slices and promotes MSC-induced angiogenesis. These beneficial effects are mediated via increases in molecules/pathways involved in tissue remodeling and angiogenesis. We speculate that E2 may enhance MSC ability to repair/regenerate cardiac tissue.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Estradiol/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Miocardio/citología , Miocardio/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/genética , Proteómica/métodos
19.
Int J Mol Sci ; 20(6)2019 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-30909560

RESUMEN

Apolipoprotein E (apoE) is mainly secreted by hepatocytes and incorporated into most plasma lipoproteins. Macrophages, which accumulate cholesterol and are critical for the development of the atherosclerotic plaque, are also an important, albeit smaller, apoE source. Distal regulatory elements control cell-specific activity of the apoE promoter: multienhancers (ME.1/2) in macrophages and hepatic control regions (HCR-1/2) in hepatocytes. A member of AP-1 cell growth regulator, c-Jun regulates the transcription of various apolipoproteins and proinflammatory molecules implicated in atherosclerosis. We aimed to investigate the effect of c-Jun on apoE expression in macrophages versus hepatocytes and to reveal the underlying molecular mechanisms. Herein we show that c-Jun had an opposite, cell-specific effect on apoE expression: downregulation in macrophages but upregulation in hepatocytes. Transient transfections using ME.2 deletion mutants and DNA pull-down (DNAP) assays showed that the inhibitory effect of c-Jun on the apoE promoter in macrophages was mediated by a functional c-Jun binding site located at 301/311 on ME.2. In hepatocytes, c-Jun overexpression strongly increased apoE expression, and this effect was due to c-Jun binding at the canonical site located at -94/-84 on the apoE proximal promoter, identified by transient transfections using apoE deletion mutants, DNAP, and chromatin immunoprecipitation assays. Overall, the dual effect of c-Jun on apoE gene expression led to decreased cholesterol efflux in macrophages resident in the atherosclerotic plaque synergized with an increased level of systemic apoE secreted by the liver to exacerbate atherogenesis.


Asunto(s)
Apolipoproteínas E/genética , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Macrófagos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Elementos de Facilitación Genéticos , Hepatocitos/inmunología , Macrófagos/inmunología , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas , Células RAW 264.7 , Factor de Transcripción AP-1/metabolismo
20.
Int J Mol Sci ; 20(24)2019 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-31842455

RESUMEN

Apolipoprotein A-I (apoA-I) is the major protein component of high-density lipoproteins (HDL), mediating many of its atheroprotective properties. Increasing data reveal the pro-atherogenic effects of bisphenol A (BPA), one of the most prevalent environmental chemicals. In this study, we investigated the mechanisms by which BPA exerts pro-atherogenic effects. For this, LDLR-/- mice were fed with a high-fat diet and treated with 50 µg BPA/kg body weight by gavage. After two months of treatment, the area of atherosclerotic lesions in the aorta, triglycerides and total cholesterol levels were significantly increased, while HDL-cholesterol was decreased in BPA-treated LDLR-/- mice as compared to control mice. Real-Time PCR data showed that BPA treatment decreased hepatic apoA-I expression. BPA downregulated the activity of the apoA-I promoter in a dose-dependent manner. This inhibitory effect was mediated by MEKK1/NF-κB signaling pathways. Transfection experiments using apoA-I promoter deletion mutants, chromatin immunoprecipitation, and protein-DNA interaction assays demonstrated that treatment of hepatocytes with BPA induced NF-κB signaling and thus the recruitment of p65/50 proteins to the multiple NF-κB binding sites located in the apoA-I promoter. In conclusion, BPA exerts pro-atherogenic effects downregulating apoA-I by MEKK1 signaling and NF-κB activation in hepatocytes.


Asunto(s)
Contaminantes Ocupacionales del Aire/efectos adversos , Apolipoproteína A-I/genética , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Compuestos de Bencidrilo/efectos adversos , Regulación de la Expresión Génica , FN-kappa B/metabolismo , Fenoles/efectos adversos , Animales , Apolipoproteína A-I/metabolismo , Aterosclerosis/sangre , Aterosclerosis/patología , Biomarcadores , Modelos Animales de Enfermedad , Femenino , Hepatocitos/metabolismo , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , Receptores de LDL/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA