Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Semin Cell Dev Biol ; 123: 22-35, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34489173

RESUMEN

Aging induces alterations in bone structure and strength through a multitude of processes, exacerbating common aging- related diseases like osteoporosis and osteoarthritis. Cellular hallmarks of aging are examined, as related to bone and the marrow microenvironment, and ways in which these might contribute to a variety of age-related perturbations in osteoblasts, osteocytes, marrow adipocytes, chondrocytes, osteoclasts, and their respective progenitors. Cellular senescence, stem cell exhaustion, mitochondrial dysfunction, epigenetic and intracellular communication changes are central pathways and recognized as associated and potentially causal in aging. We focus on these in musculoskeletal system and highlight knowledge gaps in the literature regarding cellular and tissue crosstalk in bone, cartilage, and the bone marrow niche. While senolytics have been utilized to target aging pathways, here we propose non-pharmacologic, exercise-based interventions as prospective "senolytics" against aging effects on the skeleton. Increased bone mass and delayed onset or progression of osteoporosis and osteoarthritis are some of the recognized benefits of regular exercise across the lifespan. Further investigation is needed to delineate how cellular indicators of aging manifest in bone and the marrow niche and how altered cellular and tissue crosstalk impact disease progression, as well as consideration of exercise as a therapeutic modality, as a means to enhance discovery of bone-targeted therapies.


Asunto(s)
Osteoartritis , Osteoporosis , Adipocitos , Anciano , Envejecimiento , Ejercicio Físico , Humanos , Osteoartritis/terapia , Osteoblastos , Estudios Prospectivos
2.
Cell Biochem Funct ; 42(2): e3917, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38379232

RESUMEN

A major factor in long-term impairment is stroke. Patients with persistent stroke and severe functional disabilities have few therapy choices. Long noncoding RNAs (lncRNAs) may contribute to the regulation of the pathophysiologic processes of ischemic stroke as shown by altered expression of lncRNAs and microRNA (miRNAs) in blood samples of acute ischemic stroke patients. On the other hand, multipotent mesenchymal stem cells (MSCs) increase neurogenesis, and angiogenesis, dampen neuroinflammation, and boost brain plasticity to improve functional recovery in experimental stroke models. MSCs can be procured from various sources such as the bone marrow, adipose tissue, and peripheral blood. Under the proper circumstances, MSCs can differentiate into a variety of mature cells, including neurons, astrocytes, and oligodendrocytes. Accordingly, the capability of MSCs to exert neuroprotection and also neurogenesis has recently attracted more attention. Nowadays, lncRNAs and miRNAs derived from MSCs have opened new avenues to alleviate stroke symptoms. Accordingly, in this review article, we examined various studies concerning the lncRNAs and miRNAs' role in stroke pathogenesis and delivered an overview of the therapeutic role of MSC-derived miRNAs and lncRNAs in stroke conditions.


Asunto(s)
Accidente Cerebrovascular Isquémico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , MicroARNs , ARN Largo no Codificante , Accidente Cerebrovascular , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células Madre Mesenquimatosas/metabolismo , Accidente Cerebrovascular/terapia , Accidente Cerebrovascular/metabolismo , Transducción de Señal
3.
Int J Mol Sci ; 25(10)2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38791285

RESUMEN

Extracellular vesicles (EVs) have been found to have the characteristics of their parent cells. Based on the characteristics of these EVs, various studies on disease treatment using mesenchymal stem cell (MSC)-derived EVs with regenerative activity have been actively conducted. The therapeutic nature of MSC-derived EVs has been shown in several studies, but in recent years, there have been many efforts to functionalize EVs to give them more potent therapeutic effects. Strategies for functionalizing EVs include endogenous and exogenous methods. In this study, human umbilical cord MSC (UCMSC)-derived EVs were selected for optimum OA treatments with expectation via bioinformatics analysis based on antibody array. And we created a novel nanovesicle system called the IGF-si-EV, which has the properties of both cartilage regeneration and long-term retention in the lesion site, attaching positively charged insulin-like growth factor-1 (IGF-1) to the surface of the UCMSC-derived Evs carrying siRNA, which inhibits MMP13. The downregulation of inflammation-related cytokine (MMP13, NF-kB, and IL-6) and the upregulation of cartilage-regeneration-related factors (Col2, Acan) were achieved with IGF-si-EV. Moreover, the ability of IGF-si-EV to remain in the lesion site for a long time has been proven through an ex vivo system. Collectively, the final constructed IGF-si-EV can be proposed as an effective OA treatment through its successful MMP13 inhibition, chondroprotective effect, and cartilage adhesion ability. We also believe that this EV-based nanoparticle-manufacturing technology can be applied as a platform technology for various diseases.


Asunto(s)
Vesículas Extracelulares , Factor I del Crecimiento Similar a la Insulina , Células Madre Mesenquimatosas , Osteoartritis , ARN Interferente Pequeño , Factor I del Crecimiento Similar a la Insulina/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Osteoartritis/terapia , Osteoartritis/metabolismo , ARN Interferente Pequeño/genética , Animales , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/genética
4.
Mol Cell Biochem ; 478(4): 755-765, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36083511

RESUMEN

Cardiovascular diseases, such as coronary artery disease and stroke, are the main threats to human health worldwide. Atherosclerosis, a chronic inflammatory disorder, plays a role as an initiator of all of the above-mentioned diseases. Cell therapy for diseases has attracted widespread attention. Mesenchymal stem cells (MSCs) are a type of stem cell that still exist in adults and have the characteristics of self-renewal ability, pluripotent differentiation potential, immunomodulation, tissue regeneration, anti-inflammation and low immunogenicity. In light of the properties of MSCs, some researchers have begun to target MSCs to create a possible way to alleviate atherosclerosis. Most of these studies are focused on MSC transplantation, injecting MSCs to modulate macrophages, the key inflammatory cell in atherosclerosis plaque. According to recent studies, researchers found that endothelial-to-mesenchymal transition (EndMT) has something to do with atherosclerosis development. A new cell type MSC might also appear during the EndMT process. In this article, we summarize the characteristics of MSCs, the latest progress of MSC research and its application prospects, and in view of the process of EndMT occurring in atherosclerosis, we propose some new ideas for the treatment of atherosclerosis by targeting MSCs.


Asunto(s)
Aterosclerosis , Células Progenitoras Endoteliales , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Placa Aterosclerótica , Humanos , Aterosclerosis/terapia , Aterosclerosis/metabolismo , Placa Aterosclerótica/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo
5.
Exp Cell Res ; 412(2): 113034, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35051432

RESUMEN

Tripartite motif containing 21 (TRIM21) is a member of the TRIM protein family with E3 ubiquitin ligase activity. Recent studies have demonstrated that TRIM21 widely contributes to physiological and pathological processes by ubiquitylating critical proteins in many kinds of cells. Additionally, multiple studies have shown that TRIM21 plays an important role in multiple cell differentiation processes. However, whether TRIM21 modulates the osteogenic differentiation process of mesenchymal stem cells (MSCs) remains unclear. In this study, we demonstrated that the expression of TRIM21 was decreased during the osteogenic process of MSCs and that TRIM21 negatively regulated the osteogenic capacity of MSCs both in vitro and in vivo. Moreover, we further demonstrated that TRIM21 modulated the osteogenic process of MSCs by acting as an E3 ubiquitin ligase to mediate the K48-linked ubiquitination of Akt and cause degradation. In summary, these results emphasize the critical role of TRIM21 in bone formation and TRIM21 may be a promising target to improve the clinical use of MSCs in tissue engineering.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ribonucleoproteínas/metabolismo , Ubiquitinación/fisiología , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Osteogénesis/fisiología , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Adulto Joven
6.
Molecules ; 28(5)2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36903399

RESUMEN

Mesenchymal stem cells (MSCs) have newly developed as a potential drug delivery system. MSC-based drug delivery systems (MSCs-DDS) have made significant strides in the treatment of several illnesses, as shown by a plethora of research. However, as this area of research rapidly develops, several issues with this delivery technique have emerged, most often as a result of its intrinsic limits. To increase the effectiveness and security of this system, several cutting-edge technologies are being developed concurrently. However, the advancement of MSC applicability in clinical practice is severely hampered by the absence of standardized methodologies for assessing cell safety, effectiveness, and biodistribution. In this work, the biodistribution and systemic safety of MSCs are highlighted as we assess the status of MSC-based cell therapy at this time. We also examine the underlying mechanisms of MSCs to better understand the risks of tumor initiation and propagation. Methods for MSC biodistribution are explored, as well as the pharmacokinetics and pharmacodynamics of cell therapies. We also highlight various promising technologies, such as nanotechnology, genome engineering technology, and biomimetic technology, to enhance MSC-DDS. For statistical analysis, we used analysis of variance (ANOVA), Kaplan Meier, and log-rank tests. In this work, we created a shared DDS medication distribution network using an extended enhanced optimization approach called enhanced particle swarm optimization (E-PSO). To identify the considerable untapped potential and highlight promising future research paths, we highlight the use of MSCs in gene delivery and medication, also membrane-coated MSC nanoparticles, for treatment and drug delivery.


Asunto(s)
Células Madre Mesenquimatosas , Nanopartículas , Distribución Tisular , Sistemas de Liberación de Medicamentos/métodos , Citoplasma
7.
Biochem Biophys Res Commun ; 597: 1-7, 2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-35121177

RESUMEN

INTRODUCTION: Demyelinating Charcot-Marie-Tooth disease (CMT) is caused by mutations in the genes that encode myelinating proteins or their transcription factors. Our study thus sought to assess the therapeutic effects of cytokines secreted from mesenchymal stem cells (MSCs) on this disease. METHODS: The therapeutic potential of Wharton's jelly MSCs (WJ-MSCs) and cytokines secreted by WJ-MSCs was evaluated on Schwann cells (SCs) exhibiting demyelination features, as well as a mouse model of demyelinating CMT. RESULTS: Co-culture with WJ-MSC protected PMP22-overexpressing SCs from apoptotic cell death. Using a cytokine array, the secretion of growth differentiation factor-15 (GDF-15) and amphiregulin (AREG) was found to be elevated in WJ-MSCs when co-incubated with the PMP22-overexpressing SCs. Administration of both cytokines into trembler-J (Tr-J) mice, an animal model of CMT, significantly enhanced motor nerve conduction velocity compared to the control group. More importantly, this treatment alleviated the demyelinating phenotype of Tr-J mice, as demonstrated by an improvement in the mean diameter and g-ratio of the myelinated axons. CONCLUSIONS: Our findings demonstrated that WJ-MSCs alleviate the demyelinating phenotype of CMT via the secretion of several cytokines. Further elucidation of the underlying mechanisms of GDF-15 and AREG in myelination might provide a robust basis for the development of effective therapies against demyelinating CMT.

8.
Int J Med Sci ; 18(6): 1375-1381, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33628093

RESUMEN

Introduction: Recently, the efficacy of mesenchymal stem cells (MSCs) mediated by their tissue repair and anti-inflammatory actions in the prevention and therapy of various disorders has been reported. In this research, our attention was focused specifically on the prevention and therapy of glucocorticoid-induced osteonecrosis. We investigated the stress resistance of MSC against glucocorticoid administration and hypoxic stress, which are factors known to induce osteocytic cell death. Materials and Methods: Mouse bone cells (MLO-Y4) and bone-marrow derived mouse MSCs were exposed to dexamethasone (Dex), hypoxia of 1% oxygen or both in vitro. Mitochondrial membrane potentials were estimated by mitochondria labeling with a cell-permeant probe (Mito tracker red); expression of these apoptosis-inducing molecules, oxidative stress marker (8-hydroxy-2'-deoxyguanosine), caspase-3, -9, and two apoptosis-inhibiting molecules, energy-producing ATP synthase (ATP5A) and X-linked inhibitor of apoptosis protein (XIAP), were analyzed by both immunofluorescence and western blot. Results: With exposure to either dexamethasone or hypoxia, MLO-Y4 showed reduced mitochondrial membrane potential, ATP5A and upregulation of 8-OHdG, cleaved caspases and XIAP. Those changes were significantly enhanced by treatment with dexamethasone plus hypoxia. In MSCs, however, mitochondrial membrane potentials were preserved, while no significant changes in the pro-apoptosis or anti-apoptosis molecules analyzed were found even with exposure to both dexamethasone and hypoxia. No such effects induced by treatment with dexamethasone, hypoxia, or both were demonstrated in MSCs at all. Discussion: In osteocyte cells subjected to the double stresses of glucocorticoid administration and a hypoxic environment osteocytic cell death was mediated via mitochondria. In contrast, MSC subjected to the same stressors showed preservation of mitochondrial function and reduced oxidative stress. Accordingly, even under conditions sufficiently stressful to cause the osteocytic cell death in vivo, it was thought that the function of MSC could be preserved, suggesting that in the case of osteonecrosis preventative and therapeutic strategies incorporating their intraosseous implantation may be promising.


Asunto(s)
Glucocorticoides/efectos adversos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Osteocitos/efectos de los fármacos , Osteonecrosis/terapia , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Dexametasona/efectos adversos , Modelos Animales de Enfermedad , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Osteocitos/patología , Osteonecrosis/inducido químicamente , Osteonecrosis/patología
9.
J Am Soc Nephrol ; 31(12): 2757-2772, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32753400

RESUMEN

BACKGROUND: Cell-based therapies aimed at replenishing renal parenchyma have been proposed as an approach for treating CKD. However, pathogenic mechanisms involved in CKD such as renal hypoxia result in loss of kidney function and limit engraftment and therapeutic effects of renal epithelial progenitors. Jointly administering vessel-forming cells (human mesenchymal stromal cells [MSCs] and endothelial colony-forming cells [ECFCs]) may potentially result in in vivo formation of vascular networks. METHODS: We administered renal tubule-forming cells derived from human adult and fetal kidneys (previously shown to exert a functional effect in CKD mice) into mice, alongside MSCs and ECFCs. We then assessed whether this would result in generation of "renovascular units" comprising both vessels and tubules with potential interaction. RESULTS: Directly injecting vessel-forming cells and renal tubule-forming cells into the subcutaneous and subrenal capsular space resulted in self-organization of donor-derived vascular networks that connected to host vasculature, alongside renal tubules comprising tubular epithelia of different nephron segments. Vessels derived from MSCs and ECFCs augmented in vivo tubulogenesis by the renal tubule-forming cells. In vitro coculture experiments showed that MSCs and ECFCs induced self-renewal and genes associated with mesenchymal-epithelial transition in renal tubule-forming cells, indicating paracrine effects. Notably, after renal injury, renal tubule-forming cells and vessel-forming cells infused into the renal artery did not penetrate the renal vascular network to generate vessels; only administering them into the kidney parenchyma resulted in similar generation of human renovascular units in vivo. CONCLUSIONS: Combined cell therapy of vessel-forming cells and renal tubule-forming cells aimed at alleviating renal hypoxia and enhancing tubulogenesis holds promise as the basis for new renal regenerative therapies.


Asunto(s)
Células Endoteliales/citología , Glomérulos Renales/citología , Túbulos Renales/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Técnicas de Cocultivo , Humanos , Ratones , Neovascularización Fisiológica
10.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34502137

RESUMEN

In the bone marrow of vertebrates, two types of stem cells coexist-hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). Hematopoiesis only occurs when these two stem cell types and their descendants interact. The descendants of HSCs supply the body with all the mature blood cells, while MSCs give rise to stromal cells that form a niche for HSCs and regulate the process of hematopoiesis. The studies of hematopoiesis were initially based on morphological observations, later extended by the use of physiological methods, and were subsequently augmented by massive application of sophisticated molecular techniques. The combination of these methods produced a wealth of new data on the organization and functional features of hematopoiesis in the ontogenesis of mammals and humans. This review summarizes the current views on hematopoiesis in mice and humans, discusses the development of blood elements and hematopoiesis in the embryo, and describes how the hematopoietic system works in the adult organism and how it changes during aging.


Asunto(s)
Envejecimiento/sangre , Hematopoyesis , Células Madre Hematopoyéticas/citología , Envejecimiento/metabolismo , Animales , Médula Ósea/crecimiento & desarrollo , Humanos
11.
J Transl Med ; 18(1): 241, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546185

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) have been shown to alleviate acute lung injury (ALI) and induce the production of regulatory dendritic cells (DCregs), but the potential link between these two cell types remains unclear. The goal of this study was to investigate the effect and mechanism of MSC-induced regulatory dendritic cells in ALI mice. MATERIAL/METHODS: In vivo experiments, C57BL/6 wild-type male mice were sacrificed at different times after intratracheal injection of LPS to observe changes in lung DC maturation and pathological damage. MSCs, DCregs or/and carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled DCs were administered to the mice by tail vein, and flow cytometry was performed to measure the phenotype of lung DCs and T cells. Lung injury was estimated by the lung wet weight/body weight ratio and histopathological analysis. In vitro, Western blotting or flow cytometry was used to detect the expression of Notch ligand or receptor in MSCs or DCs after coculture or LPS stimulation. Finally, in vivo and in vitro, we used the Notch signaling inhibitor DAPT to verify the effect of the Notch pathway on MSC-induced DCregs and their pulmonary protection. RESULTS: We showed significant accumulation and maturation of lung DCs 2 h after intratracheal injection of LPS, which were positively correlated with the lung pathological injury score. MSC treatment alleviated ALI lung injury, accompanied by a decrease in the number and maturity of classical DCs in the lungs. CFSE-labeled DCs migrated to the lungs of ALI mice more than those of the normal group, and the elimination of CFSE-labeled DCs in the blood was slower. MSCs inhibited the migration of CFSE-labeled DCs to the lung and promoted their elimination in the blood. DCregs, which are obtained by contact coculture of mDCs with MSCs, expressed reduced levels of MHCII, CD86, CD40 and increased levels of PD-L1, and had a reduced ability to stimulate lymphocyte proliferation and activation (expression of CD44 and CD69). mDCs expressing Notch2 significantly increased after coculture with MSCs or rhJagged1, and MSCs expressed more Jagged1 after LPS stimulation. After stimulation of mDCs with recombinant Jagged1, DCs with low expression of MHCII, CD86 and CD40 were also induced, and the effects of both rhJagged1 and MSCs on DCs were blocked by the Notch inhibitor DAPT. Intra-airway DAPT reversed the inhibitory effect of mesenchymal stem cells on DC recruitment to the lungs and its maturation. CONCLUSIONS: Our results suggested that the recruitment and maturation of lung DCs is an important process in early ALI, MSCs attenuate LPS-induced ALI by inducing the production of DCregs by activating Notch signaling.


Asunto(s)
Lesión Pulmonar Aguda , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/terapia , Animales , Células Dendríticas , Lipopolisacáridos/toxicidad , Pulmón , Masculino , Ratones , Ratones Endogámicos C57BL
12.
Dermatol Ther ; 33(6): e14277, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32888384

RESUMEN

Lichen sclerosus (LS) is a chronic relapsing, inflammatory skin disorder usually involving the anogenital region of both sexes lacking a resolutive therapy. This study compared adipose tissue derived-stromal vascular fraction (AD-SVF) and AD-SVF-enriched platelet-rich plasma (PRP) therapy in the management of genital LS patients. Additionally, in vitro evaluation of cells and growth factors contained in the injected SVF has been evaluated as possible predictive factors for treatment outcome. The study population was 40 patients diagnosed with LS who were symptomatic despite medical treatment. Patients (age 43-78 years) randomized into two groups using a 1:1 allocation ratio, were evaluated clinically and assessing dermatology life quality index (DLQI) before and 6 months after treatment. Both procedures demonstrated a strong safety profile with no complications linked to the therapy. After 6 months, both treatments allowed for a significant improvement respect to baseline. Combinatory therapy demonstrated decreased efficacy in late stage patients. No correlations have been found between clinical and biological findings. AD-SVF and AD-SVF plus PRP are safe and effective regenerative approaches for genital LS patients. Clinical results support the preferential use of combinatory therapy for early stage patients confirming a synergic effect of AD-SVF and PRP. In contrast, AD-SVF plus PRP is discouraged for late stage patients.


Asunto(s)
Liquen Escleroso y Atrófico , Tejido Adiposo , Adulto , Anciano , Femenino , Genitales , Humanos , Liquen Escleroso y Atrófico/diagnóstico , Liquen Escleroso y Atrófico/terapia , Masculino , Persona de Mediana Edad , Plasma Rico en Plaquetas , Piel
13.
Curr Osteoporos Rep ; 18(6): 774-789, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33068251

RESUMEN

PURPOSE OF REVIEW: To highlight recent basic, translational, and clinical works demonstrating exercise and diet regulation of marrow adipose tissue (MAT) and bone and how this informs current understanding of the relationship between marrow adiposity and musculoskeletal health. RECENT FINDINGS: Marrow adipocytes accumulate in the bone in the setting of not only hypercaloric intake (calorie excess; e.g., diet-induced obesity) but also with hypocaloric intake (calorie restriction; e.g., anorexia), despite the fact that these states affect bone differently. With hypercaloric intake, bone quantity is largely unaffected, whereas with hypocaloric intake, bone quantity and quality are greatly diminished. Voluntary running exercise in rodents was found to lower MAT and promote bone in eucaloric and hypercaloric states, while degrading bone in hypocaloric states, suggesting differential modulation of MAT and bone, dependent upon whole-body energy status. Energy status alters bone metabolism and bioenergetics via substrate availability or excess, which plays a key role in the response of bone and MAT to mechanical stimuli. Marrow adipose tissue (MAT) is a fat depot with a potential role in-as well as responsivity to-whole-body energy metabolism. Understanding the localized function of this depot in bone cell bioenergetics and substrate storage, principally in the exercised state, will aid to uncover putative therapeutic targets for skeletal fragility.


Asunto(s)
Tejido Adiposo/metabolismo , Médula Ósea/metabolismo , Huesos/metabolismo , Dieta , Ejercicio Físico/fisiología , Adipocitos/metabolismo , Animales , Densidad Ósea/fisiología , Metabolismo Energético/fisiología , Humanos
14.
J Cell Biochem ; 120(3): 3637-3650, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30242894

RESUMEN

Mesenchymal stem cells (MSCs) protect the endothelial barrier complex and survival, implicated in the pathogenesis of acute lung injury (ALI) via paracrine hepatocyte growth factor (HGF). However, the mechanism of HGF in endothelial regulation remains unclear. Here, we introduced a coculture protocol of pulmonary microvascular endothelial cells (PMVECs) and overexpression of the HGF gene of MSCs (MSC-HGF). Immunofluorescence and endothelial permeability analysis revealed that MSC-HGF protected endothelial tight junction protein occludin expression and attenuated cellular permeability as well as endothelial apoptosis. To investigate the novel mechanism mammalian TOR (mTOR)/ signal transducer and activator of transcription 3 (STAT-3) signaling in HGF protective effects against endothelial barrier and apoptosis, we used recombinant mouse HGF in endothelial cells. In addition, we used mTOR inhibitor rapamycin to inhibit the mTOR pathway. Our study demonstrated that rapamycin decreased the protective effects of HGF on the endothelium by decreasing tight junction protein occludin expression and cell proliferation, and raising lipopolysaccharide (LPS)-induced endothelial permeability, endothelial cell injury factors ET-1 and vWF. Similarly, the protective effects of HGF on reducing endothelial barrier and apoptosis were weakened when PMVECs were treated with the STAT-3 inhibitor S3I-201. Moreover, mTOR/STAT-3 were activated by HGF demonstrated as raising mTOR (Ser2448) and STAT3 (Ser727) phosphorylation proteins, leading to endothelial barrier improvement and survival. Reversely, rapamycin or S3I-201 inhibited mTOR/STAT-3 activation. Taken together, our findings highlight that the activation of the mTOR/STAT-3 pathway provides novel mechanistic insights into MSC-secreted HGF protection against LPS-induced vascular endothelial permeability dysfunction and apoptosis, which contributes to decreasing microvascular loss and lung injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/metabolismo , Factor de Crecimiento de Hepatocito/biosíntesis , Lipopolisacáridos/toxicidad , Células Madre Mesenquimatosas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Endoteliales/patología , Células Madre Mesenquimatosas/patología , Ratones
15.
Exp Eye Res ; 185: 107671, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31108056

RESUMEN

Through the paracrine effects of stem cells, including the secretion of neurotrophic, immunomodulatory, and anti-apoptotic factors, cell-based therapies offer a new all-encompassing approach to treatment of neurodegenerative diseases. In this study, we used physically separated co-cultures of porcine neuroretina (NR) and human mesenchymal stem cells (MSC) to evaluate the MSC paracrine neuroprotective effects on NR degeneration. NR explants were obtained from porcine eyes and cultured alone or co-cultured with commercially available MSCs from Valladolid (MSCV; Citospin S.L.; Valladolid, Spain), currently used for several approved treatments. Cultures were maintained for 72 h. MSC surface markers were evaluated before and after co-culture with NRs. Culture supernatants were collected and the concentration of brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and glial-derived neurotrophic factor (GDNF) were determined by enzyme-linked immunosorbent assays. NR sections were stained by haematoxylin/eosin or immunostained for terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL), glial fibrillary acidic protein, ß-tubulin III, and neuronal nuclei marker. NR morphology, morphometry, nuclei count, apoptosis rate, retinal ganglion cells, and glial cell activation were evaluated. Treatment effects were statistically analysed by parametric or non-parametric tests. The MSCs retained stem cell surface markers after co-culture with NR. BDNF and CNTF concentrations in NR-MSCV co-cultures were higher than other experimental conditions at 72 h (p < 0.05), but no GDNF was detected. NR general morphology, total thickness, and cell counts were broadly preserved in co-cultures, and the apoptosis rate determined by TUNEL assay was lower than for NR monocultures (all p < 0.05). Co-cultures with MSCV also protected retinal ganglion cells from degenerative changes and reduced reactive gliosis (both p < 0.05). In this in vitro model of spontaneous NR degeneration, the presence of co-cultured MSCs retarded neuroglial degeneration. This effect was associated with elevated concentrations of the neurotrophic factors BDNF and CNTF. Our data suggest that the paracrine secretion of these, and possibly other molecules, are a potential resource for the treatment of several neuroretinal diseases.


Asunto(s)
Células Madre Mesenquimatosas/citología , Neuroprotección/fisiología , Comunicación Paracrina/fisiología , Retina/citología , Degeneración Retiniana/prevención & control , Animales , Biomarcadores/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Supervivencia Celular/fisiología , Factor Neurotrófico Ciliar/metabolismo , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Células Madre Mesenquimatosas/metabolismo , Microglía/metabolismo , Retina/metabolismo , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Porcinos , Tubulina (Proteína)/metabolismo
16.
Cell Tissue Res ; 372(1): 67-75, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29082445

RESUMEN

Intravenous administration of bone marrow-derived mesenchymal stem cells (BM-MSCs) has been shown to promote nerve cell regeneration following traumatic brain injury (TBI). As the anti-oxidant defense systems in neuronal tissue including superoxide dismutase 2 (SOD2) are crucial to defend cell against oxidative stress. We proposed a new stratege to increase the therapeutic effect of MSCs by preventing cells death from oxidative stress. We overexpressed SOD2 in BM-MSCs, transplanted these MSCs into TBI model mice, assessed the protective effect of SOD2 against oxidation-induced apoptosis in BM-MSCs both in vitro and in vivo, evaluated brain functional recovery by the rotarod behavioral test, and tested the oxidation status of TBI mice brain after BM-MSCs transplantation by monitoring the superoxide dismutase, glutathione and malonaldehyde level. We found over-expression of SOD2 protected BM-MSCs from H2O2-induced cell apoptosis. Injection of SOD2 over-expressed BM-MSCs attenuated neuro-inflammation in the ipsilateral cortex of TBI mice, and protected TBI mice against loss of blood-brain barrier integrity. Furthermore, the rotarod behavioral test showed functional recovery of TBI mice after MSC treatment. Our experiments indicated that SOD2-over-expressed BM-MSCs have an improved therapeutic effect on brain injury treatment in TBI mice.


Asunto(s)
Células de la Médula Ósea/citología , Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Superóxido Dismutasa/metabolismo , Adenoviridae/metabolismo , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Citoprotección/efectos de los fármacos , Vectores Genéticos/metabolismo , Peróxido de Hidrógeno/toxicidad , Inflamación/patología , Masculino , Ratones Endogámicos BALB C , Recuperación de la Función , Transfección
17.
Exp Cell Res ; 350(2): 349-357, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-28007558

RESUMEN

The contributions of aryl hydrocarbon receptor (Ahr) to the pathogenesis of rheumatoid arthritis (RA), particularly bone loss, have not been clearly explored. The imbalance between osteoblasts and osteoclasts is a major reason for bone loss. The dysfunction of osteoblasts, which are derived from mesenchymal stem cells (MSCs), induced bone erosion occurs earlier and is characterized as more insidious. Here, we showed that the nuclear expression and translocation of Ahr were both significantly increased in MSCs from collagen-induced arthritis (CIA) mice. The enhanced Ahr suppressed the mRNA levels of osteoblastic markers including Alkaline phosphatase (Alp) and Runt-related transcription factor 2 (Runx2) in the differentiation of MSCs to osteoblasts in CIA. The 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated activation of Ahr dose-dependently suppressed the expression of osteoblastic markers. In addition, the expression of ß-catenin was reduced in CIA MSCs compared with control, and the TCDD-mediated activation of the Ahr significantly inhibited ß-catenin expression. The Wnt3a-induced the activation of Wnt/ß-catenin pathway partly rescued the osteogenesis decline induced by TCDD. Taken together, these results indicate that activated Ahr plays a negative role in CIA MSCs osteogenesis, possibly by suppressing the expression of ß-catenin.


Asunto(s)
Artritis Experimental/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Receptores de Hidrocarburo de Aril/metabolismo , beta Catenina/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Artritis Experimental/patología , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos DBA , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Dibenzodioxinas Policloradas/farmacología , Vía de Señalización Wnt
18.
Nanomedicine ; 14(7): 1987-1997, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29933024

RESUMEN

In the present study, we fabricated an efficient, simple biomimetic scaffold to stimulate osteogenic differentiation of mesenchymal stem cells (MSCs). Electrospun poly L-lactic acid nanofibers were employed to mimic the nanofibrillar structure of bone proteins and coated with hydroxyapatite nanoparticles to simulate bone minerals. Thereafter, we regulated the release pattern of BMP-2 peptide through covalent attachment of an optimized liposomal formulation to the scaffold. The fabricated platform provided a sustained release profile of BMP-2 peptide up to 21 days while supporting cellular attachment and proliferation without cytotoxicity. In-vitro results confirmed the superiority of the scaffold containing liposomes through enhancement of growth and differentiation of MSCs. Ectopic bone formation model exhibited significant localized initiation of bone formation of liposome incorporated scaffold. Consequently, these findings demonstrated that our designed platform with modified release properties of BMP-2 peptide considerably promoted osteogenic differentiation of MSCs making it a unique candidate for bone regeneration therapeutics.


Asunto(s)
Proteína Morfogenética Ósea 2/química , Regeneración Ósea , Durapatita/química , Células Madre Mesenquimatosas/citología , Nanofibras/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Ingeniería de Tejidos , Andamios del Tejido , Animales , Diferenciación Celular , Células Cultivadas , Liposomas/química , Masculino , Nanofibras/química , Fragmentos de Péptidos/química , Poliésteres/química , Ratas , Ratas Wistar
19.
Methods ; 99: 62-8, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26384580

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent stem cells. Although they were originally identified in bone marrow and described as 'marrow stromal cells', they have since been identified in many other anatomical locations in the body. MSCs can be isolated from bone marrow, adipose tissue, umbilical cord and other tissues but the richest tissue source of MSCs is fat. Since they are adherent to plastic, they may be expanded in vitro. MSCs have a distinct morphology and express a specific set of CD (cluster of differentiation) molecules. The phenotypic pattern for the identification of MSCs cells requires expression of CD73, CD90, and CD105 and lack of CD34, CD45, and HLA-DR antigens. Under appropriate micro-environmental conditions MSCs can proliferate and give rise to other cell types. Therefore, they are ideally suited for the treatment of systemic inflammatory and autoimmune conditions. They have also been implicated as key players in regenerating injured tissue following injury and trauma. MSC populations isolated from adipose tissue may also contain regulatory T (Treg) cells, which have the capacity for modulating the immune system. The immunoregulatory and regenerative properties of MSCs make them ideal for use as therapeutic agents in vivo. In this paper we review the literature on the identification, phenotypic characterization and biological properties of MSCs and discuss their potential for applications in cell therapy and regenerative medicine. We also discuss strategies for biomaterial micro-engineering of the stem cell niche.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Nicho de Células Madre , Animales , Humanos , Trasplante de Células Madre Mesenquimatosas , Fenotipo , Regeneración , Medicina Regenerativa , Ingeniería de Tejidos
20.
Methods ; 99: 69-80, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26384579

RESUMEN

Musculoskeletal disorders represent a major cause of disability and morbidity globally and result in enormous costs for health and social care systems. Development of cell-based therapies is rapidly proliferating in a number of disease areas, including musculoskeletal disorders. Novel biological therapies that can effectively treat joint and spine degeneration are high priorities in regenerative medicine. Mesenchymal stem cells (MSCs) isolated from bone marrow (BM-MSCs), adipose tissue (AD-MSCs) and umbilical cord (UC-MSCs) show considerable promise for use in cartilage and intervertebral disc (IVD) repair. This review article focuses on stem cell-based therapeutics for cartilage and IVD repair in the context of the rising global burden of musculoskeletal disorders. We discuss the biology MSCs and chondroprogenitor cells and specifically focus on umbilical cord/Wharton's jelly derived MSCs and examine their potential for regenerative applications. We also summarize key components of the molecular machinery and signaling pathways responsible for the control of chondrogenesis and explore biomimetic scaffolds and biomaterials for articular cartilage and IVD regeneration. This review explores the exciting opportunities afforded by MSCs and discusses the challenges associated with cartilage and IVD repair and regeneration. There are still many technical challenges associated with isolating, expanding, differentiating, and pre-conditioning MSCs for subsequent implantation into degenerate joints and the spine. However, the prospect of combining biomaterials and cell-based therapies that incorporate chondrocytes, chondroprogenitors and MSCs leads to the optimistic view that interdisciplinary approaches will lead to significant breakthroughs in regenerating musculoskeletal tissues, such as the joint and the spine in the near future.


Asunto(s)
Cartílago Articular/fisiología , Disco Intervertebral/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Humanos , Trasplante de Células Madre Mesenquimatosas , Regeneración , Medicina Regenerativa , Transducción de Señal , Ingeniería de Tejidos , Gelatina de Wharton/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA