Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(10)2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37239925

RESUMEN

This Special Issue focused on the complex role played by MSCs in the onset and development of inflammatory diseases: MSCs can support or counteract inflammation and, in turn, the onset of disease [...].


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Fibrosis , Inflamación , Transducción de Señal
2.
Int J Mol Sci ; 24(10)2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37239847

RESUMEN

Micro RNAs (miRNAs) are a type of non-coding RNA (ncRNA) and typically interact with specific target mRNAs through complementary base pairing, affecting their translation and/or stability. MiRNAs regulate nearly all cellular functions, including the cell fate of mesenchymal stromal cells (MSCs). It is now accepted that various pathologies arise at the stem level, and, in this scenario, the role played by miRNAs in the fate of MSCs becomes of primary concern. Here we have considered the existing literature in the field of miRNAs, MSCs and skin diseases, classified as inflammatory (such as psoriasis and atopic dermatitis-AD) and neoplastic (melanoma and non-melanoma-skin-cancer including squamous cell and basal cell carcinoma) diseases. In this scoping review article, the evidence recovered indicates that this topic has attracted attention, but it is still a matter of opinion. A protocol for this review was registered in PROSPERO with the registration number "CRD42023420245". According to the different skin disorders and to the specific cellular mechanisms considered (cancer stem cells, extracellular vesicles, inflammation), miRNAs may play a pro- or anti-inflammatory, as well as a tumor suppressive, or supporting, role, indicating a complex regulation of their function. It is evident that the mode of action of miRNAs is more than a switch on-off, and all the observed effects of their dysregulated expression must be checked in a detailed analysis of the targeted proteins. The involvement of miRNAs has been studied mainly for squamous cell carcinoma and melanoma, and much less in psoriasis and AD; different mechanisms have been considered, such as miRNAs included in extracellular vesicles derived both from MSCs or tumor cells, miRNAs involved in cancer stem cells formation, up to miRNAs as candidates to be new therapeutic tools.


Asunto(s)
Células Madre Mesenquimatosas , MicroARNs , Neoplasias , Psoriasis , Enfermedades de la Piel , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular , Enfermedades de la Piel/genética , Enfermedades de la Piel/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Psoriasis/genética , Psoriasis/metabolismo
3.
Int J Mol Sci ; 23(23)2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36499401

RESUMEN

Mesenchymal Stem Cells (MSCs) are multipotent non-hematopoietic stromal cells found in different body tissues such as bone marrow, adipose tissue, periosteum, Wharton's jelly, umbilical cord, blood, placenta, amniotic fluid, and skin. The biological behavior of MSCs depends mainly on their interaction with the microenvironment in which they are found, whose quality deeply influences the regenerative and immunomodulatory properties of these cells. Several studies confirm the interaction between MSCs and inflammatory microenvironment in the pathogenesis of psoriasis, designating MSCs as an important factor driving psoriasis development. This review aims to describe the most recent evidence on how the inflammatory microenvironment that characterizes psoriasis influences the homeostasis of MSCs and how they can be used to treat the disease.


Asunto(s)
Células Madre Mesenquimatosas , Gelatina de Wharton , Embarazo , Femenino , Humanos , Diferenciación Celular , Cordón Umbilical , Líquido Amniótico
4.
Inflamm Res ; 70(1): 79-87, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33210178

RESUMEN

INTRODUCTION: Psoriasis cellular hallmarks, such as the imbalance between Th1/Th17 and Th2 cytokines and the dysregulated expression of vascular endothelial growth factor (VEGF), inducible nitric oxide synthase, (iNOS) and indoleamine 2,3-dioxygenase (IDO), are all detectable in mesenchymal stem cells (MSCs) suggesting that psoriasis originates at mesenchymal level. AIM OF THE STUDY: In this scenario, MSCs may become the new therapeutic target and interest in the effects of traditionally used drugs, such as Apremilast, on MSCs has greatly increased. MATERIALS AND METHODS: MSCs from control subjects (C-MSCs) and from psoriatic patients before (PsO MSCs T0) and after in vivo treatment with Apremilast (PsO-MSCs T12) were isolated and characterized; subsequently, the effects of Apremilast on VEGF, iNOS and IDO expression were evaluated by immunocytochemistry (ICC). The expression of VEGF, iNOS and IDO was also detected in skin sections by immunohistochemistry (IHC). RESULTS: The results indicate that in vivo administration of Apremilast is able to drive the altered profile of PsO-MSCs towards a more physiological pattern. In skin sections, the role of Apremilast is evident in reducing VEGF, iNOS and IDO expression. CONCLUSION: Apremilast treatment influences the expression of VEGF, iNOS and IDO not only by keratinocytes but also by MSCs, restoring their intrinsic profile and their natural anti-inflammatory action, and decreasing the auto-inflammatory process that underpins the development of psoriasis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Psoriasis/inmunología , Talidomida/análogos & derivados , Adulto , Anciano , Antiinflamatorios no Esteroideos/uso terapéutico , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Masculino , Células Madre Mesenquimatosas/inmunología , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo II/inmunología , Psoriasis/tratamiento farmacológico , Piel/efectos de los fármacos , Piel/inmunología , Talidomida/farmacología , Talidomida/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/inmunología
5.
Mol Biol Rep ; 48(8): 5935-5942, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34319544

RESUMEN

BACKGROUND: Male infertility is a problem that affects 10-15% of men of reproductive age. In particular, gametogenesis is a complex process in which inflammation may play a central role through the secretion of cytokines and the expression of microRNAs. We assessed the potential role of proinflammatory cytokines (TNF-α, IL-6 and IL-1α) and microRNAs (miR-146a-5p, miR-34a-5p and miR-23a-3p) in the seminal plasma of infertile men compared to controls, evaluating their correlation with seminal and biochemical parameters. METHODS AND RESULTS: Expression of cytokines and microRNAs was analyzed by ELISA and q-PCR. Our data shows that IL-1α was significantly increased in the azoospermic group compared to controls, TNF-α mRNA was more expressed in the oligozoospermic group than controls. There were no significant differences in miRNAs expression among the three groups. The correlations between sperm parameters and inflammatory markers were evaluated, however no significance was highlighted. CONCLUSIONS: The determination of each inflammatory marker separately in the seminal plasma of subfertile men, despite some significant differences, does not have a diagnostic value in male infertility even if an assay of selective pro-inflammatory cytokines and microRNAs in the semen may improve the diagnosis of male infertility.


Asunto(s)
Infertilidad Masculina/genética , Infertilidad Masculina/inmunología , Semen/fisiología , Adulto , Biomarcadores/metabolismo , Citocinas/metabolismo , Humanos , Interleucina-1alfa/metabolismo , Interleucina-6/metabolismo , Masculino , MicroARNs/genética , Semen/metabolismo , Espermatozoides/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Túnez
6.
Am J Obstet Gynecol ; 222(6): 596.e1-596.e9, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31874141

RESUMEN

BACKGROUND: Uterine leiomyomas (fibroids) are smooth muscle neoplasms of the myometrial layer of the uterus and are the most common benign tumors in women. Although their etiology is still unclear, progenitor cells seem to be implicated. OBJECTIVE: To identify the dysregulated pathways involved in leiomyoma onset by microRNA profiling of progenitor cells isolated from normal myometrium and leiomyoma tissue. MATERIALS AND METHODS: Pairs of normal myometrium and uterine fibroid specimens were collected from 12 myomectomy patients. Myometrial progenitor cells and leiomyoma progenitor cells were isolated and characterized for stemness. After total RNA extraction and profiling of their 2646 microRNAs, DIANA-miRPath analysis was applied to find any dysregulated pathways. RESULTS: Only 30 microRNAs showed a significant differential regulation between myometrial progenitor cells and leiomyoma progenitor cells. Removal of those that had values close to the cut-off or that were not consistent among triplicates left 15 microRNAs, of which 7 were downregulated and 8 were upregulated in leiomyoma progenitor cells compared to myometrial progenitor cells. According to DIANA-miRPath analysis, the 7 downregulated microRNAs (hsa-miR-146b-5p; hsa-miR-335-3p; hsa-miR-335-5p; hsa-miR-135b-5p; hsa-miR-10a-3p; hsa-miR-10a-5p; hsa-miR-200a-3p) are all related to 3 pathways, "ECM-receptor interaction" (33 targeted genes), "Adherens junction" (33 targeted genes), and "Hippo signaling" (69 targeted genes), whereas the 8 upregulated miRNAs (hsa-miR-146a-5p; hsa-miR-576-3p; hsa-miR-122-5p; hsa-miR-1246; hsa-miR-595; hsa-miR-658; hsa-miR-4284; hsa-miR-924) are related to 4 pathways, "PI3K-Akt signaling pathway" (71 targeted genes), "Pathways in Cancer" (80 targeted genes), "Cell Cycle" (37 targeted genes), and "Regulation of actin cytoskeleton" (41 targeted genes). CONCLUSION: The findings that only 15 of 2646 microRNAs are differentially regulated in normal myometrium and leiomyoma and that they are involved in 7 dysregulated pathways provides interesting insights into the development of uterine fibroids, and lends support to the hypothesis that leiomyoma onset is the result of alterations affecting progenitor cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Leiomioma/genética , MicroARNs/genética , Miometrio/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre/metabolismo , Neoplasias Uterinas/genética , Citoesqueleto de Actina/genética , Uniones Adherentes/genética , Adulto , Ciclo Celular/genética , Regulación hacia Abajo , Matriz Extracelular/genética , Femenino , Humanos , Leiomioma/metabolismo , Leiomioma/cirugía , Miometrio/citología , Transducción de Señal/genética , Regulación hacia Arriba , Miomectomía Uterina , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/cirugía , Población Blanca/genética
7.
Int J Mol Sci ; 21(21)2020 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-33143370

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic disease characterized by fibroblasts activation, ECM accumulation, and diffused alveolar inflammation. The role of inflammation in IPF is still controversial and its involvement may follow nontraditional mechanisms. It is seen that a pathological microenvironment may affect cells, in particular mesenchymal stem cells (MSCs) that may be able to sustain the inflamed microenvironment and influence the surrounding cells. Here MSCs have been isolated from fibrotic (IPF-MSCs) and control (C-MSCs) lung tissue; first cells were characterized and compared by the expression of molecules related to ECM, inflammation, and other interdependent pathways such as hypoxia and oxidative stress. Subsequently, MSCs were co-cultured between them and with NHLF to test the effects of the cellular crosstalk. Results showed that pathological microenvironment modified the features of MSCs: IPF-MSCs, compared to C-MSCs, express higher level of molecules related to ECM, inflammation, oxidative stress, and hypoxia; notably, when co-cultured with C-MSCs and NHLF, IPF-MSCs are able to induce a pathological phenotype on the surrounding cell types. In conclusion, in IPF the pathological microenvironment affects MSCs that in turn can modulate the behavior of other cell types favoring the progression of IPF.


Asunto(s)
Biomarcadores/metabolismo , Microambiente Celular , Fibroblastos/patología , Fibrosis Pulmonar Idiopática/patología , Inflamación/patología , Pulmón/patología , Células Madre Mesenquimatosas/patología , Anciano , Biomarcadores/análisis , Estudios de Casos y Controles , Proliferación Celular , Técnicas de Cocultivo , Femenino , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Inflamación/metabolismo , Pulmón/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo
8.
Med Microbiol Immunol ; 208(6): 877-883, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31214759

RESUMEN

In the past few years the increasing incidence of hospital infections with Acinetobacter baumannii, especially in immunocompromised patients, and its proneness to develop multidrug resistance have been raising considerable concern. This study examines the antimicrobial and antibiofilm activity of protegrin 1 (PG-1), an antimicrobial peptide from porcine leukocytes, against A. baumannii strains isolated from surgical wounds. PG-1 was tested both alone and combined with the antibiotics commonly used in clinical settings. Its antimicrobial activity was evaluated by determination of minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC), checkerboard assays, and time-kill experiments. Its effects on biofilm inhibition/eradication were tested with crystal violet staining. The strains were grown in subinhibitory or increasing PG-1 concentrations to test the development of resistance. Mammalian cell toxicity was tested by XTT assays. PG-1 MICs and MBCs ranged from 2 to 8 µg/ml. PG-1 was most active and demonstrated a synergistic interaction with colistin, a last resort antibiotic. Interestingly, antagonism was never observed. In time-kill experiments, incubation with 2 × MIC for 30 min suppressed all viable cells. PG-1 did not select resistant strains and showed a limited effect on cell viability, but it did exert a strong activity against multidrug-resistant A. baumannii. In contrast, in our experimental conditions it had no effect on biofilm inhibition/eradication. PG-1 thus seems to be a promising antimicrobial agent against multidrug-resistant Gram-negative infections.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antiinfecciosos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas/efectos de los fármacos , Interacciones Farmacológicas , Herida Quirúrgica/microbiología , Acinetobacter baumannii/aislamiento & purificación , Antiinfecciosos/toxicidad , Péptidos Catiónicos Antimicrobianos/toxicidad , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana , Coloración y Etiquetado
9.
J Cell Physiol ; 233(6): 4423-4442, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29159853

RESUMEN

Tissue regenerative medicine combines the use of cells, scaffolds, and molecules to repair damaged tissues. Different cell types are employed for musculoskeletal diseases, both differentiated and mesenchymal stromal cells (MSCs). In recent years, the hypothesis that cell-based therapy is guided principally by cell-secreted factors has become increasingly popular. The aim of the present literature review was to evaluate preclinical and clinical studies that used conditioned medium (CM), rich in cell-factors, for musculoskeletal regeneration. Thirty-one were in vitro, 12 in vivo studies, 1 was a clinical study, and 2 regarded extracellular vesicles. Both differentiated cells and MSCs produce CM that induces reduction in inflammation and increases synthetic activity. MSC recruitment and differentiation, endothelial cell recruitment and angiogenesis have also been observed. In vivo studies were performed with CM in bone and periodontal defects, arthritis and muscle dystrophy pathologies. The only clinical study was performed with CM from MSCs in patients needing alveolar bone regeneration, showing bone formation and no systemic or local complications. Platelet derived growth factor receptor ß, C3a, vascular endothelial growth factor, monocyte chemoattractant protein-1 and -3, interleukin 3 and 6, insulin-like growth factor-I were identified as responsible of cell migration, proliferation, osteogenic differentiation, and angiogenesis. The use of CM could represent a new regenerative treatment in several musculoskeletal pathologies because it overcomes problems associated with the use of cells and avoids the use of exogenous GFs or gene delivery systems. However, some issues remain to be clarified.


Asunto(s)
Terapia Biológica/métodos , Medios de Cultivo Condicionados/metabolismo , Células Madre Mesenquimatosas/metabolismo , Enfermedades Musculoesqueléticas/terapia , Sistema Musculoesquelético/metabolismo , Comunicación Paracrina , Regeneración , Medicina Regenerativa/métodos , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Enfermedades Musculoesqueléticas/metabolismo , Enfermedades Musculoesqueléticas/fisiopatología , Sistema Musculoesquelético/fisiopatología , Fenotipo , Transducción de Señal
10.
Exp Dermatol ; 26(4): 319-324, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27376466

RESUMEN

Psoriasis is a disease characterized by an imbalance between Th1 and Th17 and Th2 inflammatory axes, in which cutaneous mesenchymal stem cells (MSCs) are early involved, as they show a greater relative expression of several genes encoding for Th1 and Th17 cytokines. Therapeutic implications of TNF-α inhibitors on differentiated skin cells have been largely described in psoriasis; however, their effects on MSCs derived from patients with psoriasis have been only partially described. The aim of this work was to evaluate the effect of TNF-α inhibitors on cytokine milieu expressed by MSCs isolated from the skin of patients with psoriasis. Resident MSCs from skin of patients with psoriasis and healthy subjects have been isolated, characterized and profiled by PCR and ELISA for the expression of 22 cytokines involved in Th1 , Th2 and Th17 pathways, both before and after 12 weeks therapy with TNF-α inhibitors. The administration of TNF-α inhibitors for 12-weeks acts on MSCs as follows: it reduces the expression of several Th1 -Th17 cytokines whose levels are elevated at baseline (IL-6, IL-8, IL-12, IL-23A, IFN-γ, TNF-α, CCL2, CCL20, CXCL2, CXCL5, IL-17A, IL-17C, IL-17F, IL-21, G-CSF). Similarly, it enhances the expression of several Th2 cytokines which are underexpressed at baseline (IL-2, IL-4, IL-5), reducing the expression of those overexpressed at baseline (TGF-ß and IL-13). TNF-α inhibitors could contribute to reduce the pathological imbalance between the Th1 -Th17 vs Th2 axis in MSCs of patients with psoriasis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Citocinas/genética , Células Madre Mesenquimatosas/metabolismo , Psoriasis/tratamiento farmacológico , Psoriasis/fisiopatología , Linfocitos T Colaboradores-Inductores/metabolismo , Adalimumab/uso terapéutico , Estudios de Casos y Controles , Células Cultivadas , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Etanercept/uso terapéutico , Expresión Génica/efectos de los fármacos , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/inmunología , Estudios Prospectivos , ARN Mensajero/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA