Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
Front Bioeng Biotechnol ; 11: 1189225, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37229487

RESUMEN

Maintenance of tissue homeostasis and tissue regeneration after an insult are essential functions of adult stem cells (SCs). In adult tissues, SCs proliferate at a very slow rate within "stem cell niches", but, during tissue development and regeneration, before giving rise to differentiated cells, they give rise to multipotent and highly proliferative cells, known as transit-amplifying cells (TACs). Although differences exist in diverse tissues, TACs are not only a transitory phase from SCs to post-mitotic cells, but they also actively control proliferation and number of their ancestor SCs and proliferation and differentiation of their progeny toward tissue specific functional cells. Autocrine signals and negative and positive feedback and feedforward paracrine signals play a major role in these controls. In the present review we will consider the generation and the role played by TACs during development and regeneration of lining epithelia characterized by a high turnover including epidermis and hair follicles, ocular epithelial surfaces, and intestinal mucosa. A comparison between these different tissues will be made. There are some genes and molecular pathways whose expression and activation are common to most TACs regardless their tissue of origin. These include, among others, Wnt, Notch, Hedgehog and BMP pathways. However, the response to these molecular signals can vary in TACs of different tissues. Secondly, we will consider cultured cells derived from tissues of mesodermal origin and widely adopted for cell therapy treatments. These include mesenchymal stem cells and dedifferentiated chondrocytes. The possible correlation between cell dedifferentiation and reversion to a transit amplifying cell stage will be discussed.

2.
Front Bioeng Biotechnol ; 10: 869408, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35586557

RESUMEN

Cutaneous chronic wounds are a major global health burden in continuous growth, because of population aging and the higher incidence of chronic diseases, such as diabetes. Different treatments have been proposed: biological, surgical, and physical. However, most of these treatments are palliative and none of them can be considered fully satisfactory. During a spontaneous wound healing, endogenous regeneration mechanisms and resident cell activity are triggered by the released platelet content. Activated stem and progenitor cells are key factors for ulcer healing, and they can be either recruited to the wound site from the tissue itself (resident cells) or from elsewhere. Transplant of skin substitutes, and of stem cells derived from tissues such as bone marrow or adipose tissue, together with platelet-rich plasma (PRP) treatments have been proposed as therapeutic options, and they represent the today most promising tools to promote ulcer healing in diabetes. Although stem cells can directly participate to skin repair, they primarily contribute to the tissue remodeling by releasing biomolecules and microvesicles able to stimulate the endogenous regeneration mechanisms. Stem cells and PRP can be obtained from patients as autologous preparations. However, in the diabetic condition, poor cell number, reduced cell activity or impaired PRP efficacy may limit their use. Administration of allogeneic preparations from healthy and/or younger donors is regarded with increasing interest to overcome such limitation. This review summarizes the results obtained when these innovative treatments were adopted in preclinical animal models of diabetes and in diabetic patients, with a focus on allogeneic preparations.

4.
Int J Mol Sci ; 21(14)2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32698534

RESUMEN

To understand the regenerative effect of platelet-released molecules in bone repair one should investigate the cascade of events involving the resident osteoblast population during the reconstructive process. Here the in vitro response of human osteoblasts to a platelet lysate (PL) stimulus is reported. Quiescent or very slow dividing osteoblasts showed a burst of proliferation after PL stimulation and returned to a none or very slow dividing condition when the PL was removed. PL stimulated osteoblasts maintained a differentiation capability in vitro and in vivo when tested in absence of PL. Since angiogenesis plays a crucial role in the bone healing process, we investigated in PL stimulated osteoblasts the activation of hypoxia-inducible factor 1-alpha (HIF-1α) and signal transducer and activator of transcription 3 (STAT3) pathways, involved in both angiogenesis and bone regeneration. We observed phosphorylation of STAT3 and a strong induction, nuclear translocation and DNA binding of HIF-1α. In agreement with the induction of HIF-1α an enhanced secretion of vascular endothelial growth factor (VEGF) occurred. The double effect of the PL on quiescent osteoblasts, i.e., resumption of proliferation and activation of pathways promoting both angiogenesis and bone formation, provides a rationale to the application of PL as therapeutic agent in post-traumatic bone repair.


Asunto(s)
Plaquetas/metabolismo , Regeneración Ósea , Huesos/irrigación sanguínea , Huesos/lesiones , Neovascularización Fisiológica , Osteoblastos/citología , Adulto , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Osteoblastos/metabolismo , Osteogénesis , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Cells ; 9(4)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32340136

RESUMEN

Regenerative strategies for human articular cartilage are still challenging despite the presence of resident progenitor cell population. Today, many efforts in the field of regenerative medicine focus on the use of platelet derivatives due to their ability to reactivate endogenous mechanisms supporting tissue repair. While their use in orthopedics continues, mechanisms of action and efficacy need further characterization. We describe that the platelet lysate (PL) is able to activate chondro-progenitor cells in a terminally differentiated cartilage tissue. Primary cultures of human articular chondrocytes (ACs) and cartilage explants were set up from donor hip joint biopsies and were treated in vitro with PL. PL recruited a chondro-progenitors (CPCs)-enriched population from ex vivo cartilage culture, that showed high proliferation rate, clonogenicity and nestin expression. CPCs were positive for in vitro tri-lineage differentiation and formed hyaline cartilage-like tissue in vivo without hypertrophic fate. Moreover, the secretory profile of CPCs was analyzed, together with their migratory capabilities. Some CPC-features were also induced in PL-treated ACs compared to fetal bovine serum (FBS)-control ACs. PL treatment of human articular cartilage activates a stem cell niche responsive to injury. These facts can improve the PL therapeutic efficacy in cartilage applications.


Asunto(s)
Plaquetas/metabolismo , Cartílago Articular/citología , Cartílago Articular/fisiología , Regeneración/fisiología , Ingeniería de Tejidos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Senescencia Celular , Condrogénesis , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Hipertrofia , Inflamación/patología , Ratones Desnudos , Persona de Mediana Edad , Nestina/metabolismo , Fenotipo , Células Madre/metabolismo
6.
Pharmaceutics ; 12(2)2020 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-32028579

RESUMEN

Standard treatments of chronic skin ulcers based on the direct application of dressings still present several limits with regard to a complete tissue regeneration. Innovative strategies in tissue engineering offer materials that can tune cell behavior and promote growth tissue favoring cell recruitment in the early stages of wound healing. A combination of Alginate (Alg), Sericin (SS) with Platelet Lysate (PL), as a freeze-dried sponge, is proposed to generate a bioactive wound dressing to care skin lesions. Biomembranes at different composition were tested for the release of platelet growth factors, cytotoxicity, protective effects against oxidative stress and cell proliferation induction. The highest level of the growth factors release occurred within 48 h, an optimized time to burst a healing process in vivo; the presence of SS differently modulated the release of the factors by interaction with the proteins composing the biomembranes. Any cytotoxicity was registered, whereas a capability to protect cells against oxidative stress and induce proliferation was observed when PL was included in the biomembrane. In a mouse skin lesion model, the biomembranes with PL promoted the healing process, inducing an accelerated and more pronounced burst of inflammation, formation of granulation tissue and new collagen deposition, leading to a more rapid skin regeneration.

7.
In Vivo ; 33(6): 1851-1855, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31662512

RESUMEN

AIM: To develop a method capable of identifying human corneal limbal stem cells (LSCs) and follow their proliferation and migration in the epithelium. MATERIALS AND METHODS: Ten fresh matched pairs of cadaveric normal human corneas were obtained from donors. Carboxyfluorescein diacetate succinimidyl ester (CFSE) was used to target LSCs. The distribution of CFSE-positive cell clusters was analyzed by fluorescence microscopy by counterstaining with 4',6-diamidino-2-phenylindole (DAPI). Fluorescence was digitally recorded for seven days, and the rate of cell movement was determined. RESULTS: CFSE-labeled cells were tracked in corneas. Analysis of time sequences revealed that they moved centripetally. Daily average CFSE-labeled LSC movement was 0.073±0.01 cm (±SD). CONCLUSION: CFSE allowed us to identify LSCs and to track their centripetal migration from the limbal basal layer to the anterior ocular surface. This experimental system appears to be a valuable tool for further studies on corneal epithelial cell migration and proliferation.


Asunto(s)
Movimiento Celular/fisiología , Córnea/fisiología , Epitelio Corneal/fisiología , Fluoresceínas/metabolismo , Células Madre/fisiología , Succinimidas/metabolismo , Técnicas de Cultivo de Célula/métodos , Proliferación Celular/fisiología , Córnea/metabolismo , Epitelio Corneal/metabolismo , Humanos , Células Madre/metabolismo
8.
Cells ; 8(4)2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30970613

RESUMEN

: Injured blood vessel repair and blood circulation re-establishment are crucial events for tissue repair. We investigated in primary cultures of human umbilical vein endothelial cells (HUVEC), the effects of platelet lysate (PL), a cocktail of factors released by activated platelets following blood vessel disruption and involved in the wound-healing process triggering. PL exerted a protective effect on HUVEC in an inflammatory milieu by inhibiting IL-1α-activated NF-κB pathway and by inducing the secretion of PGE2, a pro-resolving molecule in the wound microenvironment. Moreover, PL enhanced HUVEC proliferation, without affecting their capability of forming tube-like structures on matrigel, and activated resting quiescent cells to re-enter cell cycle. In agreement with these findings, proliferation-related pathways Akt and ERK1/2 were activated. The expression of the cell-cycle activator Cyclin D1 was also enhanced, as well as the expression of the High Mobility Group Box-1 (HMGB1), a protein of the alarmin group involved in tissue homeostasis, repair, and remodeling. These in vitro data suggest a possible in vivo contribution of PL to new vessel formation after a wound by activation of cells resident in vessel walls. Our biochemical study provides a rationale for the clinical use of PL in the treatment of wound healing-related pathologies.


Asunto(s)
Factores de Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Plaquetas/citología , Diferenciación Celular , Células Cultivadas , Ciclina D1/metabolismo , Proteína HMGB1/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
9.
Acta Biomater ; 89: 33-46, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30880235

RESUMEN

X-ray Synchrotron radiation-based techniques, in particular Micro-tomography and Micro-diffraction, were exploited to investigate the structure of bone deposited in vivo within a porous ceramic scaffold. Bone formation was studied by implanting Mesenchymal Stem Cell (MSC) seeded ceramic scaffolds in a mouse model. Osteoblasts derived from the seeded MSC and from differentiation of cells migrated within the scaffold together with the blood vessels, deposited within the scaffold pores an organic collagenous matrix on which a precursor mineral amorphous liquid-phase, containing Ca++ and PO4-- crystallized filling the gaps between the collagen molecules. Histology offered a valid instrument to investigate the engineered tissue structure, but, unfortunately, limited itself to a macroscopic analysis. The evolution of the X-ray Synchrotron radiation-based techniques and the combination of micro X-ray diffraction with X-ray phase-contrast imaging enabled to study the dynamic of the structural and morphological changes occurring during the new bone deposition, biomineralization and vascularization. In fact, the unique features of Synchrotron radiation, is providing the high spatial resolution probe which is necessary for the study of complex materials presenting heterogeneity from micron-scale to meso- and nano-scale. Indeed, this is the occurrence in the heterogeneous and hierarchical bone tissue where an organic matter, such as the collagenous matrix, interacts with mineral nano-crystals to generate a hybrid multiscale biomaterial with unique physical properties. In this framework, the use of advanced synchrotron radiation techniques allowed to understand and to clarify fundamental aspects of the bone formation process within the bioceramic, i.e. biomineralization and vascularization, including to obtain deeper knowledge on bone deposition, mineralization and reabsorption in different health, aging and pathological conditions. In this review we present an overview of the X-ray Synchrotron radiation techniques and we provide a general outlook of their applications on bone Tissue Engineering, with a focus on our group work. STATEMENT OF SIGNIFICANCE: Synchrotron Radiation techniques for Tissue Engineering In this review we report recent applications of X-ray Synchrotron radiation-based techniques, in particular Microtomography and Microdiffraction, to investigations on the structure of ceramic scaffolds and bone tissue regeneration. Tissue engineering has made significant advances in bone regeneration by proposing the use of mesenchymal stem cells in combination with various types of scaffolds. The efficacy of the biomaterials used to date is not considered optimal in terms of resorbability and bone formation, resulting in a poor vascularization at the implant site. The review largely based on our publications in the last ten years could help the study of the regenerative model proposed. We also believe that the new imaging technologies we describe could be a starting point for the development of additional new techniques with the final aim of transferring them to the clinical practice.


Asunto(s)
Huesos/metabolismo , Diferenciación Celular , Cerámica/química , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Sincrotrones , Ingeniería de Tejidos , Andamios del Tejido/química , Difracción de Rayos X , Animales , Calcificación Fisiológica , Ratones , Porosidad
10.
J Tissue Eng Regen Med ; 13(6): 1007-1018, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30811859

RESUMEN

Due to their osteoconductive and inductive properties, a variety of calcium phosphate (CaP) scaffolds are commonly used in orthopaedics as graft material to heal bone defects. In this study, we have used two CaP scaffolds with different hydroxyapatite (HA) and ß-tricalcium phosphate (ß-TCP) ratios (MBCP®; 60/40 and MBCP+ ®; 20/80) to investigate their intrinsic capacity to favour human bone marrow stem cells (hBMSCs) osteogenic differentiation capacity. We report that MBCP+ ® showed in in vitro culture model a higher rate of calcium ion release in comparison with MBCP®. In two defined coculture systems, the hBMSC seeded onto MBCP+ ® presented an increased amount of VEGF secretion, resulting in an enhanced endothelial cell proliferation and capillary formation compared with hBMSC seeded onto MBCP®. When both ceramics combined with hBMSC were implanted in a nude mouse model, we observed a faster osteogenic differentiation and enhancement mature bone deposition sustained by the presence of a vast host vasculature within the MBCP+ ® ceramics. Bone formation was observed in samples highly positive to the activation of calcium sensing receptor protein (CaSr) on the surface of seeded hBMSC that also shown higher BMP-2 protein expression. With these data we provide valuable insights in the possible mechanisms of ossification and angiogenesis by hBMSC that we believe to be primed by calcium ions released from CaP scaffolds. Evidences could lead to an optimization of ceramic scaffolds to prime bone repair.


Asunto(s)
Fosfatos de Calcio/farmacología , Cerámica/farmacología , Células Madre Mesenquimatosas/citología , Osteogénesis/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Durapatita/química , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Receptores Sensibles al Calcio/metabolismo , Transducción de Señal , Ingeniería de Tejidos
11.
Stem Cell Res Ther ; 9(1): 300, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30409222

RESUMEN

BACKGROUND: Restoration of damaged tissues through the activation of endogenous progenitors is an attractive therapeutic option. A deep evaluation of the intrinsic stem/progenitor cell properties as well as the reciprocal interactions with injured environments is of critical importance. METHODS: Here, we show that bone marrow stromal cell antigen 2 (BST2) allows the isolation of a population of circulating progenitors, the circulating healing (CH) cells, characterized by a distinctive core signature. The bone marrow (BM) origin of BST2pos CH cells has been strengthened by the co-expression of leptin receptor, the hallmark of a subpopulation of BM-skeletal stem cells. RESULTS: BST2pos CH cells retained the capacity to (i) respond to injury signals generated by a bone fracture, (ii) modify the expression of cell motility genes following damage, and (iii) react to hepatocyte growth factor-activator (HGFA), an injury-related stimulus sufficient to induce their transition into GALERT, a state in which cells are functionally activated and participate in tissue repair. CONCLUSIONS: Taken together, these results could pave the way for the identification of new strategies to enhance and potentiate endogenous regenerative mechanisms for future therapies.


Asunto(s)
Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Glicoproteínas de Membrana/metabolismo , Serina Endopeptidasas/farmacología , Cicatrización de Heridas , Heridas y Lesiones/patología , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Cicatrización de Heridas/efectos de los fármacos
12.
Acta Biomater ; 73: 365-376, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29673841

RESUMEN

Human platelet lysate (hPL) is a pool of growth factors and cytokines able to induce regeneration of different tissues. Despite its good potentiality as therapeutic tool for regenerative medicine applications, hPL has been only moderately exploited in this field. A more widespread adoption has been limited because of its rapid degradation at room temperature that decreases its functionality. Another limiting factor for its extensive use is the difficulty of handling the hPL gels. In this work, silk fibroin-based patches were developed to address several points: improving the handling of hPL, enabling their delivery in a controlled manner and facilitating their storage by creating a device ready to use with expanded shelf life. Patches of fibroin loaded with hPL were synthesized by electrospinning to take advantage of the fibrous morphology. The release kinetics of the material was characterized and tuned through the control of fibroin crystallinity. Cell viability assays, performed with primary human dermal fibroblasts, demonstrated that fibroin is able to preserve the hPL biological activity and prolong its shelf-life. The strategy of storing and preserving small active molecules within a naturally-derived, protein-based fibrous scaffold was successfully implemented, leading to the design of a biocompatible device, which can potentially simplify the storage and the application of the hPL on a human patient, undergoing medical procedures such as surgery and wound care. STATEMENT OF SIGNIFICANCE: Human platelets lysate (hPL) is a mixture of growth factors and cytokines able to induce the regeneration of damaged tissues. This study aims at enclosing hPL in a silk fibroin electrospun matrix to expand its utilization. Silk fibroin showed the ability to preserve the hPL activity at temperature up to 60 °C and the manipulation of fibroin's crystallinity provided a tool to modulate the hPL release kinetic. This entails the possibility to fabricate the hPL silk fibroin patches in advance and store them, resulting in an easy and fast accessibility and an expanded use of hPL for wound healing.


Asunto(s)
Plaquetas/metabolismo , Sistemas de Liberación de Medicamentos , Fibroínas/química , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Animales , Materiales Biocompatibles , Bombyx , Citocinas/metabolismo , Preparaciones de Acción Retardada , Fibroblastos/efectos de los fármacos , Humanos , Cinética , Microscopía Confocal , Microscopía Electrónica de Rastreo , Nanofibras , Estructura Secundaria de Proteína , Regeneración , Temperatura , Agua/química , Cicatrización de Heridas
13.
Front Physiol ; 9: 181, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593553

RESUMEN

Bone remodeling process consists in a slow building phase and in faster resorption with the objective to maintain a functional skeleton locomotion to counteract the Earth gravity. Thus, during spaceflights, the skeleton does not act against gravity, with a rapid decrease of bone mass and density, favoring bone fracture. Several studies approached the problem by imaging the bone architecture and density of cosmonauts returned by the different spaceflights. However, the weaknesses of the previously reported studies was two-fold: on the one hand the research suffered the small statistical sample size of almost all human spaceflight studies, on the other the results were not fully reliable, mainly due to the fact that the observed bone structures were small compared with the spatial resolution of the available imaging devices. The recent advances in high-resolution X-ray tomography have stimulated the study of weight-bearing skeletal sites by novel approaches, mainly based on the use of the mouse and its various strains as an animal model, and sometimes taking advantage of the synchrotron radiation support to approach studies of 3D bone architecture and mineralization degree mapping at different hierarchical levels. Here we report the first, to our knowledge, systematic review of the recent advances in studying the skeletal bone architecture by high-resolution X-ray tomography after submission of mice models to microgravity constrains.

14.
Artículo en Inglés | MEDLINE | ID: mdl-30622945

RESUMEN

Skin chronic wounds are non-healing ulcerative defects, which arise in association with a morbidity state, such as diabetes and vascular insufficiency or as the consequence of systemic factors including advanced age. Platelet Rich Plasma, a platelet-rich blood fraction, can significantly improve the healing of human skin chronic ulcers. Given that the subcutaneous adipose tissue is located beneath the skin and plays a role in the skin homeostasis, in this study, we investigated the in vitro response of human subcutaneous adipose tissue cells to platelet content in a model mimicking in vitro the in situ milieu of a deep skin injury. Considering that, at the wound site, plasma turn to serum, platelets are activated and inflammation occurs, human adipose-derived stromal cells (hASC) were cultured with Human Serum (HS) supplemented or not with Platelet Lysate (PL) and/or IL-1α. We observed that HS sustained hASC proliferation more efficiently than FBS and induced a spontaneous adipogenic differentiation in the cells. PL added to HS enhanced hASC proliferation, regardless the presence of IL-1α. In the presence of PL, hASC progressively lessened the adipogenic phenotype, possibly because the proliferation of less committed cells was induced. However, these cells resumed adipogenesis in permissive conditions. Accordingly, PL induced in quiescent cells activation of the proliferation-related pathways ERK, Akt, and STAT-3 and expression of Cyclin D1. Moreover, PL induced an early and transient increase of the pro-inflammatory response triggered by IL-1α, by inducing COX-2 expression and secretion of a large amount of PGE2, IL-6, and IL-8. Media conditioned by PL-stimulated hASC exerted a chemotactic activity on human keratinocytes and favored the healing of an in vitro scratch wound. In order to bridge the gap between in vitro results and possible in vivo events, the stimuli were also tested in ex vivo cultures of in toto human adipose tissue biopsies (hAT). PL induced cell proliferation in hAT and outgrowth of committed progenitor cells able to differentiate in permissive conditions. In conclusion, we report that the adipose tissue responds to the wound microenvironment by activating the proliferation of adipose tissue progenitor cells and promoting the release of factors favoring wound healing.

15.
J Tissue Eng Regen Med ; 12(1): e82-e96, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-27863057

RESUMEN

Chronic skin ulcers, consequence of diabetes and other pathological conditions, heavily compromise the patient life quality and represent a high and constantly growing cost for National Health Services. Autologous platelet-rich plasma (PRP), has been proposed to treat these lesions. The absence of guidelines for the PRP production and the need of a fresh preparation for each treatment lead us to develop a protocol for the production of an allogenic PRP-based bioactive membrane (BAM), standardized for platelet concentration and growth factor release. This work compares BAMs obtained starting from two different platelet concentrations. There was no direct correlation between the amount of growth factors released by BAM in vitro and the initial platelet count. However, different release kinetics were noticed for different growth factors, suggesting that they were differently retained by the two BAMs. The angiogenic potential of both BAMs was determined by Luminex Angiogenesis Assay. The biological activity of the factors released by the two BAMs was confirmed by cell proliferation and migration. A diabetic mouse chronic ulcer model was used to define the best PRP therapeutic dose in vivo. Both BAMs induced wound healing by increasing the thickness of the regenerated epidermis and the vessel number. However, a too high platelet concentration resulted in a slowdown of the membrane resorption that interfered with the skin healing. Overall, the results indicate that the BAMs could represent a natural and effective wound healing tool for the treatment of skin ulcers. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Materiales Biocompatibles/farmacología , Membranas Artificiales , Plasma Rico en Plaquetas/metabolismo , Cicatrización de Heridas , Animales , Plaquetas/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cinética , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Úlcera Cutánea/patología
16.
J Tissue Eng Regen Med ; 12(1): 30-43, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-27863082

RESUMEN

Autologous platelet-rich plasma (PRP) is used clinically to induce repair of different tissues through the release of bioactive molecules. In some patients, the production of efficient autologous PRP is unfeasible due to their compromised health. Allogeneic PRP mismatched for AB0 and Rh antigens was developed. The effect of allogeneic PRP on immune response should be defined to use it in clinical practice avoiding side effects. Thus, whether PRP affects the differentiation of peripheral blood monocytes to dendritic cells upon stimulation with granulocyte monocyte colony stimulating factor and interleukin-4 was investigated. Indeed, these cells are the main players of immune response and tissue repair. PRP inhibited the differentiation of monocytes to CD1a+ dendritic cells and favoured the expansion of phagocytic CD163+ CD206+ fibrocyte-like cells. These cells produced interleukin-10 and prostaglandin-E2 , but not interferon-γ, upon stimulation with lipopolysaccharides. Moreover, they promoted the expansion of regulatory CD4+ CD25+ FoxP3+ T cells upon allostimulation or antigen specific priming. Finally, the conditioned medium harvested from monocytes differentiated with PRP triggered a strong chemotactic effect on mesenchymal cells in both scratch and transwell migration assays. These results strongly suggest that allogeneic PRP can foster the differentiation of monocytes to a regulatory anti-inflammatory population, possibly favouring wound healing. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Microambiente Celular , Células Dendríticas/citología , Monocitos/citología , Plasma Rico en Plaquetas/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Antiinflamatorios/farmacología , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Dinoprostona/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Humanos , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Prueba de Cultivo Mixto de Linfocitos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Fagocitos/citología , Fagocitos/efectos de los fármacos , Fenotipo , Toxina Tetánica/farmacología , Trasplante Homólogo , Levaduras/efectos de los fármacos , Levaduras/metabolismo
17.
J Tissue Eng Regen Med ; 12(3): e1691-e1703, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29052350

RESUMEN

The idea of rescuing the body self-repair capability lost during evolution is progressively gaining ground in regenerative medicine. In particular, growth factors and bioactive molecules derived from activated platelets emerged as promising therapeutic agents acting as trigger for repair of tissue lesions and restoration of tissue functions. Aim of this study was to assess the potential of a platelet lysate (PL) for human articular cartilage repair considering its activity on progenitor cells and differentiated chondrocytes. PL induced the re-entry in the cell cycle of confluent, growth-arrested dedifferentiated/progenitor cartilage cells. In a cartilage permissive culture environment, differentiated cells also resumed proliferation after exposure to PL. These findings correlated with an up-regulation of the proliferation/survival pathways ERKs and Akt and with an induction of cyclin D1. In short- and long-term cultures of articular cartilage explants, we observed a release of proliferating chondroprogenitors able to differentiate and form an "in vitro" tissue with properties of healthy articular cartilage. Moreover, in cultured cartilage cells, PL induced a hypoxia-inducible factor (HIF-1) alpha increase, its nuclear relocation and the binding to HIF-1 responsive elements. These events were possibly related to the cell proliferation because the HIF-1 inhibitor acriflavine inhibited HIF-1 binding to HIF-1 responsive elements and cell proliferation. Our study demonstrates that PL induces quiescent cartilage cell activation and proliferation leading to new cartilage formation, identifies PL activated pathways playing a role in these processes, and provides a rationale to the application of PL for therapeutic treatment of damaged articular cartilage.


Asunto(s)
Plaquetas/química , Cartílago Articular/citología , Reprogramación Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Anciano , Anciano de 80 o más Años , Cartílago/metabolismo , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Condrocitos/citología , Matriz Extracelular/metabolismo , Humanos , Persona de Mediana Edad
18.
Artículo en Inglés | MEDLINE | ID: mdl-29209609

RESUMEN

Present cell culture medium supplements, in most cases based on animal sera, are not fully satisfactory especially for the in vitro expansion of cells intended for human cell therapy. This paper refers to (i) an heparin-free human platelet lysate (PL) devoid of serum or plasma components (v-PL) and (ii) an heparin-free human serum derived from plasma devoid of PL components (Pl-s) and to their use as single components or in combination in primary or cell line cultures. Human mesenchymal stem cells (MSC) primary cultures were obtained from adipose tissue, bone marrow, and umbilical cord. Human chondrocytes were obtained from articular cartilage biopsies. In general, MSC expanded in the presence of Pl-s alone showed a low or no proliferation in comparison to cells grown with the combination of Pl-s and v-PL. Confluent, growth-arrested cells, either human MSC or human articular chondrocytes, treated with v-PL resumed proliferation, whereas control cultures, not supplemented with v-PL, remained quiescent and did not proliferate. Interestingly, signal transduction pathways distinctive of proliferation were activated also in cells treated with v-PL in the absence of serum, when cell proliferation did not occur, indicating that v-PL could induce the cell re-entry in the cell cycle (cell commitment), but the presence of serum proteins was an absolute requirement for cell proliferation to happen. Indeed, Pl-s alone supported cell growth in constitutively activated cell lines (U-937, HeLa, HaCaT, and V-79) regardless of the co-presence of v-PL. Plasma- and plasma-derived serum were equally able to sustain cell proliferation although, for cells cultured in adhesion, the Pl-s was more efficient than the plasma from which it was derived. In conclusion, the cells expanded in the presence of the new additives maintained their differentiation potential and did not show alterations in their karyotype.

19.
Regen Med ; 12(5): 525-539, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28770657

RESUMEN

AIM: This study aimed to identify host cell recruitment patterns in a mouse model in response to rhBMP-2 releasing hyaluronic acid hydrogels and influence of added nano-hydroxyapatite particles on rhBMP-2 release and pattern of bone formation. MATERIALS & METHODS: Implanted gels were retrieved after implantation and cells were enzymatically dissociated for flow cytometric analysis. Percentages of macrophages, progenitor endothelial cells and putative mesenchymal stem cells were measured. Implants were evaluated for BMP-2 release by ELISA and by histology to monitor tissue formation. RESULTS & CONCLUSION: Hyaluronic acid+BMP-2 gels influenced the inflammatory response in the bone healing microenvironment. Host-derived putative mesenchymal stem cells were major contributors. Addition of hydroxyapatite nanoparticles modified the release pattern of rhBMP-2, resulting in enhanced bone formation.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Ácido Hialurónico/química , Hidrogeles/química , Tejido Subcutáneo/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Preparaciones de Acción Retardada , Durapatita/química , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Humanos , Implantes Experimentales , Inflamación/patología , Cinética , Macrófagos/citología , Macrófagos/efectos de los fármacos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Nanopartículas/química , Osteogénesis/efectos de los fármacos , Proteínas Recombinantes/farmacología
20.
Artículo en Inglés | MEDLINE | ID: mdl-28503549

RESUMEN

For repair of chronic or difficult-to-heal tissue lesions and defects, major constraints exist to a broad application of cell therapy and tissue engineering approaches, i.e., transplantation of "ex vivo" expanded autologous stem/progenitor cells, alone or associated with carrier biomaterials. To enable a large number of patients to benefit, new strategies should be considered. One of the main goals of contemporary regenerative medicine is to develop new regenerative therapies, inspired from Mother Nature. In all injured tissues, when platelets are activated by tissue contact, their released factors promote innate immune cell migration to the wound site. Platelet-derived factors and factors secreted by migrating immune cells create an inflammatory microenvironment, in turn, causing the activation of angiogenesis and vasculogenesis processes. Eventually, repair or regeneration of the injured tissue occurs via paracrine signals activating, mobilizing or recruiting to the wound site cells with healing potential, such as stem cells, progenitors, or undifferentiated cells derived from the reprogramming of tissue differentiated cells. This review, largely based on our studies, discusses the identification of new tools, inspired by cellular and molecular mechanisms overseeing physiological tissue healing, that could reactivate dormant endogenous regeneration mechanisms lost during evolution and ontogenesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...