Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-39302089

RESUMEN

PURPOSE: This systematic review aimed to investigate in animal models the presence of disease-modifying effects driven by non-bone marrow-derived and non-adipose-derived products, with a particular focus on umbilical cord and placenta-derived cell-based therapies for the intra-articular injective treatment of osteoarthritis (OA). METHODS: A systematic review was performed on three electronic databases (PubMed, Web of Science and Embase) according to PRISMA guidelines. The results were synthesised to investigate disease-modifying effects in preclinical animal studies comparing injectable umbilical cord, placenta, and other sources-derived products with OA controls. The risk of bias was assessed using the SYRCLE tool. RESULTS: A total of 80 studies were included (2314 animals). Cell therapies were most commonly obtained from the umbilical cord in 33 studies and placenta/amniotic tissue in 18. Cell products were xenogeneic in 61 studies and allogeneic in the remaining 19 studies. Overall, 25/27 (92.6%) of studies on umbilical cord-derived products documented better results compared to OA controls in at least one of the following outcomes: macroscopic, histological and/or immunohistochemical findings, with 19/22 of studies (83.4%) show positive results at the cartilage level and 4/6 of studies (66.7%) at the synovial level. Placenta-derived injectable products documented positive results in 13/16 (81.3%) of the studies, 12/15 (80.0%) at the cartilage level, and 2/4 (50.0%) at the synovial level, but 2/16 studies (12.5%) found overall worse results than OA controls. Other sources (embryonic, synovial, peripheral blood, dental pulp, cartilage, meniscus and muscle-derived products) were investigated in fewer preclinical studies. The risk of bias was low in 42% of items, unclear in 49%, and high in 9% of items. CONCLUSION: Interest in cell-based injectable therapies for OA treatment is soaring, particularly for alternatives to bone marrow and adipose tissue. While expanded umbilical cord mesenchymal stem cells reported auspicious disease-modifying effects in preventing OA progression in animal models, placenta/amniotic tissue also reported deleterious effects on OA joints. Lower evidence has been found for other cellular sources such as embryonic, synovial, peripheral blood, dental-pulp, cartilage, meniscus, and muscle-derived products. LEVEL OF EVIDENCE: Level II.

2.
Front Cell Dev Biol ; 12: 1456416, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39234562

RESUMEN

Renal fibrosis, characterized by excessive extracellular matrix accumulation, leads to a progressive decline of renal function and is a common endpoint of chronic kidney disease (CKD). Current treatments primarily focus on managing underlying diseases, offering limited direct intervention for the fibrotic process. This study explores the anti-fibrotic potential of human adipose-derived mesenchymal stromal cells (MSCs) and their derived extracellular vesicles (EVs) in the context of CKD, emphasizing the effects of systemic versus local delivery methods. Preconditioned MSCs (Pr-MSCs) were treated with TNF-α and IFN-γ to enhance their immunomodulatory capabilities, and demonstrated significant anti-fibrotic effects in vitro, reducing mRNA expression of fibrosis markers in TGF-ß stimulated HKC-8 cells. Our in vivo findings from a murine unilateral ureteral obstruction (UUO) model of CKD showed that local deliveries of Pr-MSCs reduced collagen deposition and increased expression of the anti-inflammatory cytokine IL-10. Systemic administration of Pr-MSCs did not show any significant effect on UUO-induced injury. In addition, EVs did not replicate the anti-fibrotic effects observed with their parent cells, suggesting that soluble proteins or metabolites secreted by Pr-MSCs might be the primary mediators of the anti-fibrotic and immunomodulatory effects. This study provides critical insights into the therapeutic efficacy of MSCs, highlighting the importance of delivery methods and the potential of preconditioning strategies in enhancing MSC-based therapies for renal fibrosis.

3.
Cells ; 13(15)2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39120301

RESUMEN

Head and neck cancer (HNC) entails a heterogenous neoplastic disease that arises from the mucosal epithelium of the upper respiratory system and the gastrointestinal tract. It is characterized by high morbidity and mortality, being the eighth most common cancer worldwide. It is believed that the mesenchymal/stem stromal cells (MSCs) present in the tumour milieu play a key role in the modulation of tumour initiation, development and patient outcomes; they also influence the resistance to cisplatin-based chemotherapy, the gold standard for advanced HNC. MSCs are multipotent, heterogeneous and mobile cells. Although no MSC-specific markers exist, they can be recognized based on several others, such as CD73, CD90 and CD105, while lacking the presence of CD45, CD34, CD14 or CD11b, CD79α, or CD19 and HLA-DR antigens; they share phenotypic similarity with stromal cells and their capacity to differentiate into other cell types. In the tumour niche, MSC populations are characterized by cell quiescence, self-renewal capacity, low reactive oxygen species production and the acquisition of epithelial-to-mesenchymal transition properties. They may play a key role in the process of acquiring drug resistance and thus in treatment failure. The present narrative review examines the links between MSCs and HNC, as well as the different mechanisms involved in the development of resistance to current chemo-radiotherapies in HNC. It also examines the possibilities of pharmacological targeting of stemness-related chemoresistance in HNSCC. It describes promising new strategies to optimize chemoradiotherapy, with the potential to personalize patient treatment approaches, and highlights future therapeutic perspectives in HNC.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias de Cabeza y Cuello , Células Madre Mesenquimatosas , Humanos , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/terapia , Neoplasias de Cabeza y Cuello/inmunología , Células Madre Mesenquimatosas/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Carcinogénesis/patología , Carcinogénesis/efectos de los fármacos , Animales , Trasplante de Células Madre Mesenquimatosas
4.
Biomedicines ; 12(6)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38927412

RESUMEN

The periosteum plays a critical role in bone repair and is significantly influenced by the surrounding immune microenvironment. In this study, we employed 10× single-cell RNA sequencing to create a detailed cellular atlas of the swine cranial periosteum, highlighting the cellular dynamics and interactions essential for cranial bone injury repair. We noted that such injuries lead to an increase in M2 macrophages, which are key in modulating the periosteum's immune response and driving the bone regeneration process. These macrophages actively recruit periosteal stromal cells (PSCs) by secreting Neuregulin 1 (NRG1), a crucial factor in initiating bone regeneration. This recruitment process emphasizes the critical role of PSCs in effective bone repair, positioning them as primary targets for therapeutic interventions. Our results indicate that enhancing the interaction between M2 macrophages and PSCs could significantly improve the outcomes of treatments aimed at cranial bone repair and regeneration.

5.
Front Immunol ; 15: 1405249, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38742110

RESUMEN

Introduction: Exploring monocytes' roles within the tumor microenvironment is crucial for crafting targeted cancer treatments. Methods: This study unveils a novel methodology utilizing four 20-color flow cytometry panels for comprehensive peripheral immune system phenotyping, specifically targeting classical, intermediate, and non-classical monocyte subsets. Results: By applying advanced dimensionality reduction techniques like t-distributed stochastic neighbor embedding (tSNE) and FlowSom analysis, we performed an extensive profiling of monocytes, assessing 50 unique cell surface markers related to a wide range of immunological functions, including activation, differentiation, and immune checkpoint regulation. Discussion: This in-depth approach significantly refines the identification of monocyte subsets, directly supporting the development of personalized immunotherapies and enhancing diagnostic precision. Our pioneering panel for monocyte phenotyping marks a substantial leap in understanding monocyte biology, with profound implications for the accuracy of disease diagnostics and the success of checkpoint-inhibitor therapies. Key findings include revealing distinct marker expression patterns linked to tumor progression and providing new avenues for targeted therapeutic interventions.


Asunto(s)
Biomarcadores , Citometría de Flujo , Inmunofenotipificación , Monocitos , Humanos , Monocitos/inmunología , Monocitos/metabolismo , Citometría de Flujo/métodos , Análisis por Conglomerados , Inmunofenotipificación/métodos , Microambiente Tumoral/inmunología , Neoplasias/inmunología , Neoplasias/diagnóstico
6.
Int J Mol Sci ; 25(4)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38397098

RESUMEN

Multipotent mesenchymal stromal cells (MSCs) integrate hormone and neuromediator signaling to coordinate tissue homeostasis, tissue renewal and regeneration. To facilitate the investigation of MSC biology, stable immortalized cell lines are created (e.g., commercially available ASC52telo). However, the ASC52telo cell line has an impaired adipogenic ability and a depressed response to hormones, including 5-HT, GABA, glutamate, noradrenaline, PTH and insulin compared to primary cells. This markedly reduces the potential of the ASC52telo cell line in studying the mechanisms of hormonal control of MSC's physiology. Here, we have established a novel immortalized culture of adipose tissue-derived MSCs via forced telomerase expression after lentiviral transduction. These immortalized cell cultures demonstrate high proliferative potential (up to 40 passages), delayed senescence, as well as preserved primary culture-like functional activity (sensitivity to hormones, ability to hormonal sensitization and differentiation) and immunophenotype up to 17-26 passages. Meanwhile, primary adipose tissue-derived MSCs usually irreversibly lose their properties by 8-10 passages. Observed characteristics of reported immortalized human MSC cultures make them a feasible model for studying molecular mechanisms, which regulate the functional activities of these cells, especially when primary cultures or commercially available cell lines are not appropriate.


Asunto(s)
Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/metabolismo , Línea Celular , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Hormonas/metabolismo , Proliferación Celular
7.
Odontology ; 112(3): 938-949, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38367068

RESUMEN

Medication-related osteonecrosis of the jaw (MRONJ) is an intractable adverse event. Dental implants are one of the triggering factors of MRONJ, and implant therapy with low MRONJ risk is required. This study aimed to investigate a rat model of MRONJ induced by extraoral placement of titanium materials and the use of mesenchymal stromal cell (MSCs) sheets to prevent MRONJ. Eight-week-old male rats were administered zoledronate and dexamethasone thrice weekly until killing. A week after drug initiation, a titanium screw and a plate were placed on the left buccal side of the mandible. Allogeneic bone marrow-derived MSC sheets were co-grafted with the titanium plates in the MSC sheet ( +) group. Six weeks after titanium placement, the rats were killed, and their excised mandibular bones were subjected to micro-computed tomography (CT) analysis. Histological analysis was performed after the titanium implants were removed. Empty lacunae visualized on hematoxylin and eosin staining were used as evidence of bone necrosis. Bone necrosis was reduced in the MSC sheet ( +) group. Tartrate-resistant acid phosphatase (TRAP) staining revealed a decreased number of TRAP-positive cells in areas with a large number of empty lacunae in the MSC sheet (-) group. Micro-CT analyses demonstrated that the bone volume fraction (BV/TV) was not significantly different between the MSC sheet (-) and ( +) groups. We conclude that MRONJ can be triggered by a titanium placement in rats, and grafting of allogeneic MSC sheets has the potential to prevent MRONJ.


Asunto(s)
Osteonecrosis de los Maxilares Asociada a Difosfonatos , Implantes Dentales , Titanio , Microtomografía por Rayos X , Ácido Zoledrónico , Animales , Ratas , Masculino , Osteonecrosis de los Maxilares Asociada a Difosfonatos/prevención & control , Osteonecrosis de los Maxilares Asociada a Difosfonatos/etiología , Implantes Dentales/efectos adversos , Dexametasona , Trasplante de Células Madre Mesenquimatosas , Imidazoles , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Difosfonatos/efectos adversos , Mandíbula/cirugía , Conservadores de la Densidad Ósea/efectos adversos
8.
Stem Cell Res Ther ; 14(1): 363, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38087388

RESUMEN

BACKGROUND: Despite a long history of investigation and sustained efforts in clinical testing, the number of market authorisations for mesenchymal stromal cell (MSC) therapies remains limited, with none approved by the United States Food and Drug Administration. Several barriers are impeding the clinical progression of MSC therapies, to the forefront of these is a lack of standardised manufacturing protocols which is further compounded by an absence of biologically meaningful characterisation and release assays. A look at clinical trial registries demonstrates the diversity of MSC expansion protocols with variabilities in cell source, isolation method and expansion medium, among other culture variables, making it extraordinarily difficult to compare study outcomes. Current identification and characterisation standards are insufficient; they are not specific to MSCs and do not indicate cell function or therapeutic action. METHODS: This work analysed the influence of five widely used culture media formulations on the colony-forming potential, proliferation kinetics, trilineage differentiation potential and immunomodulatory potential of human bone marrow-derived MSCs (BM-MSCs). The surface marker expression profiles were also characterised using a high-content flow cytometry screening panel of 243 markers. RESULTS: Significant differences in the biological attributes of BM-MSCs including clonogenicity, proliferation, differentiation propensity and immunomodulatory capacity were revealed in response to the composition of the culture medium. Despite their biological differences, all cell preparations uniformly and strongly expressed the standard positive markers proposed for BM-MSCs: CD73, CD90 and CD105. Immunophenotypic profiling revealed that the culture medium also had a significant influence on the surface proteome, with one-third of tested markers exhibiting variable expression profiles. Principal component analysis demonstrated that BM-MSCs isolated and expanded in a proprietary xeno- and serum-free medium displayed the most consistent cell phenotypes with little variability between donors compared to platelet lysate and foetal bovine serum-containing media. CONCLUSIONS: These data suggest that media composition has a highly significant impact on the biological attributes of MSCs, but standard surface marker tests conceal these differences. The results indicate a need for (1) standardised approaches to manufacturing, with an essential focus on defined media and (2) new biologically relevant tests for MSC characterisation and product release.


Asunto(s)
Células Madre Mesenquimatosas , Humanos , Proliferación Celular , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular , Citometría de Flujo , Fenotipo , Células de la Médula Ósea , Células Cultivadas , Medios de Cultivo/farmacología , Medios de Cultivo/metabolismo
9.
Cells ; 12(24)2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38132108

RESUMEN

Mesenchymal stromal cells (MSCs) have recently been shown to play an important role in the growth and progression of many solid tumors, including cholangiocarcinoma (CCA). The human placental amniotic membrane (hPAM) is one of the most favorable sources of MSCs due to its availability and non-invasive harvesting procedure. However, the role of human placental amniotic membrane mesenchymal stromal cells (hPAMSCs) in the growth and progression of human CCA has not yet been determined. This study investigates the effects of conditioned medium derived from hPAMSCs (PA-CM) on the properties of three human CCA cell lines and explores possible mechanisms of action. Varying concentrations of PA-CM were used to treat CCA cells to determine their effects on the proliferation and apoptosis of CCA cells. The results showed that PA-CM inhibited the proliferation and colony-forming capacity of KKU100, KKU213A, and KKU213B cells. PA-CM also promoted the apoptosis of these CCA cells by causing the loss of mitochondrial membrane potential. Western Blotting confirmed that PA-CM induced CCA cell apoptosis by increasing the levels of the Bax/Bcl-2 ratio, cleaved caspase 3, and cleaved PARP, possibly by inhibiting the IL-6/JAK2/STAT3 signaling pathway. Moreover, our in vivo study also confirmed the suppressive effect of hPAMSCs on CCA cells by showing that PA-CM reduced tumor volume in nude mice transplanted with human CCA cells. Taken together, our results demonstrate that PA-CM has potent tumor-suppressive effects on human CCA cells and could potentially be used in combination with chemotherapy to develop a more effective treatment for CCA patients.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Células Madre Mesenquimatosas , Embarazo , Animales , Ratones , Humanos , Femenino , Interleucina-6/metabolismo , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/metabolismo , Amnios/metabolismo , Ratones Desnudos , Proliferación Celular , Placenta/metabolismo , Colangiocarcinoma/patología , Transducción de Señal , Conductos Biliares Intrahepáticos/patología , Neoplasias de los Conductos Biliares/patología , Apoptosis , Células Madre Mesenquimatosas/metabolismo , Janus Quinasa 2/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-37815189

RESUMEN

Wound infection often requires a long period of care and an onerous treatment process. Also, the rich environment makes the wound an ideal niche for microbial growth. Stable structures, like biofilm, and drug-resistant strains cause a delay in the healing process, which has become one of the important challenges in wound treatment. Many studies have focused on alternative methods to deal the wound infections. One of the novel and highly potential ways is mesenchymal stromal cells (MSCs). MSCs are mesoderm-derived pluripotent adult stem cells with the capacity for self-renewal, multidirectional differentiation, and immunological control. Also, MSCs have anti-inflammatory and antiapoptotic effects. MScs, as pluripotent stromal cells, differentiate into many mature cells. Also, MSCs produce antimicrobial compounds, such as antimicrobial peptides (AMP), as well as secrete immune modulators, which are two basic features considered in wound healing. Despite the advantages, preserving the structure and activity of MSCs is considered one of the most important points in the treatment. MSCs' antimicrobial effects on microorganisms involved in wound infection have been confirmed in various studies. In this review, we aimed to discuss the antimicrobial and therapeutic applications of MSCs in the infected wound healing processes.

11.
Stem Cell Res Ther ; 14(1): 289, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37798772

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) have a dynamic secretome that plays a critical role in tissue repair and regeneration. However, studying the MSC secretome in mixed-culture disease models remains challenging. This study aimed to develop a mutant methionyl-tRNA synthetase-based toolkit (MetRSL274G) to selectively profile secreted proteins from MSCs in mixed-culture systems and demonstrate its potential for investigating MSC responses to pathological stimulation. METHODS: We used CRISPR/Cas9 homology-directed repair to stably integrate MetRSL274G into cells, enabling the incorporation of the non-canonical amino acid, azidonorleucine (ANL), and facilitating selective protein isolation using click chemistry. MetRSL274G was integrated into both in H4 cells and induced pluripotent stem cells (iPSCs) for a series of proof-of-concept studies. Following iPSC differentiation into induced-MSCs, we validated their identity and co-cultured MetRSL274G-expressing iMSCs with naïve or lipopolysaccharide (LPS)-treated THP-1 cells. We then profiled the iMSC secretome using antibody arrays. RESULTS: Our results showed successful integration of MetRSL274G into targeted cells, allowing specific isolation of proteins from mixed-culture environments. We also demonstrated that the secretome of MetRSL274G-expressing iMSCs can be differentiated from that of THP-1 cells in co-culture and is altered when co-cultured with LPS-treated THP-1 cells compared to naïve THP-1 cells. CONCLUSIONS: The MetRSL274G-based toolkit we have generated enables selective profiling of the MSC secretome in mixed-culture disease models. This approach has broad applications for examining not only MSC responses to models of pathological conditions, but any other cell type that can be differentiated from iPSCs. This can potentially reveal novel MSC-mediated repair mechanisms and advancing our understanding of tissue regeneration processes.


Asunto(s)
Células Madre Mesenquimatosas , Metionina-ARNt Ligasa , Metionina-ARNt Ligasa/genética , Metionina-ARNt Ligasa/metabolismo , Lipopolisacáridos , Secretoma , Células Madre Mesenquimatosas/metabolismo , Aminoácidos
12.
Dokl Biochem Biophys ; 510(1): 95-98, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37582870

RESUMEN

The osteogenic potential of mesenchymal stromal cells (MSCs) can determine bone homeostasis and the physical characteristics of bones. Microgravity reduces the ability of these cells to differentiate in osteogenic direction. It has been shown that the addition of hematopoietic stem and progenitor cells (HSPCs) to MSC culture in vitro can have the opposite effect. The aim of this study was to identify transcriptional changes in 84 genes associated with Wnt signaling in MSCs during microgravity simulation and interaction with HSPCs. The results indicate an increase in the non-canonical Wnt signaling activity during coculturing of MSCs and HSPCs, while simulated microgravity enhances the canonical component of this signaling pathway. These changes may underlie the modulation of osteogenic potential of MSCs in hematopoietic niche under microgravity.


Asunto(s)
Células Madre Mesenquimatosas , Ingravidez , Vía de Señalización Wnt/genética , Diferenciación Celular , Técnicas de Cocultivo , Osteogénesis , Células Cultivadas
13.
Stem Cell Res Ther ; 14(1): 187, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507751

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) have been shown to exert their therapeutic effects through the secretion of broad spectrum of paracrine factors, including extracellular vesicles (EVs). Accordingly, EVs are being pursued as a promising alternative to cell-based therapies. Menstrual blood-derived stromal cells (MenSCs) are a type of MSC that, due to their immunomodulatory and regenerative properties, have emerged as an innovative source. Additionally, new strategies of cell priming may potentially alter the concentration and cargo of released EVs, leading to modification of their biological properties. In this study, we aimed to characterize the EVs released by MenSCs and compare their therapeutic potential under three different preconditioning conditions (proinflammatory stimuli, physioxia, and acute hypoxia). METHODS: MenSCs were isolated from five healthy women. Following culturing to 80% confluence, MenSCs were exposed to different priming conditions: basal (21% O2), proinflammatory stimuli (IFNγ and TNFα, 21% O2), physioxia (1-2% O2), and acute hypoxia (< 1% O2) for 48-72 h. Conditioned media from MenSCs was collected after 48 h and EVs were isolated by a combination of ultra-filtration and differential centrifugation. An extensive characterization ranging from nano-flow cytometry (nFC) to quantitative high-throughput shotgun proteomics was performed. Bioinformatics analyses were used to derive hypotheses on their biological properties. RESULTS: No differences in the morphology, size, or number of EVs released were detected between priming conditions. The proteome analysis associated with basal MenSC-EVs prominently revealed their immunomodulatory and regenerative capabilities. Furthermore, quantitative proteomic analysis of differentially produced MenSC-EVs provided sufficient evidence for the utility of the differential preconditioning in purpose-tailoring EVs for their therapeutic application: proinflammatory priming enhanced the anti-inflammatory, regenerative and immunomodulatory capacity in the innate response of EVs, physioxia priming also improves tissue regeneration, angiogenesis and their immunomodulatory capacity targeting on the adaptive response, while acute hypoxia priming, increased hemostasis and apoptotic processes regulation in MenSC-EVs, also by stimulating immunomodulation mainly through the adaptive response. CONCLUSIONS: Priming of MenSCs under proinflammatory and hypoxic conditions affected the cargo proteome of EVs released, resulting in different therapeutic potential, and thus warrants experimental exploration with the aim to generate better-defined MSC-derived bioproducts.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Humanos , Femenino , Proteómica , Proteoma , Hipoxia/terapia
14.
J Orthop Translat ; 40: 104-115, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37457311

RESUMEN

Objective: To explore the repair effect of tissue engineering for annulus fibrosus (AF) injury in stress-stimulation environment. Methods: Non-adhesive fibrinogen (Fib) representing the repair with non-stress stimulation and adhesive hydrogel of fibrinogen, thrombin and genipin mixture (Fib-T-G) representing the repair with stress stimulation were prepared to repair the AF lesion. The relationship between adhesion and stress stimulation was studied in rheological measurements, tension tests and atomic force microscopy (AFM) experiments. The repair effect of stress stimulation was studied in designed acellular AF scaffold models with fissures and defects. The models were repaired by the two different hydrogels, then implanted subcutaneously and cultured for 21 â€‹d in rats. Histology and qPCR of COL1A1, COL2A1, aggrecan, RhoA, and ROCK of the tissue engineering of the interface were evaluated afterward. Moreover, the repair effect was also studied in an AF fissure model in caudal disc of rats by the two different hydrogels. Discs were harvested after 21 â€‹d, and the disc degeneration score and AF healing quality were evaluated by histology. Result: In interfacial stress experiment, Fib-T-G hydrogel showed greater viscosity than Fib hydrogel (24.67 â€‹± â€‹1.007 vs 459333 â€‹± â€‹169205 â€‹mPa â€‹s). Representative force-displacement and sample modulus for each group demonstrate that Fib-T-G group significantly increased the interfacial stress level and enhanced the modulus of samples, compared with Fib group (P â€‹< â€‹0.01). The Fib-T-G group could better bond the interface to resist the loading strain force with the broken point at 1.11 â€‹± â€‹0.10 â€‹N compared to the Fib group at 0.12 â€‹± â€‹0.08 â€‹N â€‹(P â€‹< â€‹0.01). Focusing on the interfacial healing in acellular AF scaffold model, compared with Fib â€‹+ â€‹MSCs group, the fissure and defect were connected closely in Fib-T-G â€‹+ â€‹MSCs group (P â€‹< â€‹0.01). Relative higher gene expression of COL2A1 and RhoA in Fib-T-G â€‹+ â€‹MSCs group than Fib â€‹+ â€‹MSCs group in AF fissure and AF defect model (P â€‹< â€‹0.05). The immunohistochemistry staining showed more positive staining of COL2A1 and RhoA in Fib-T-G â€‹+ â€‹MSCs group than in Fib â€‹+ â€‹MSCs group in both AF fissure and AF defect models. The degree of disc degeneration was more severe in Fib â€‹+ â€‹MSCs group than Fib-T-G â€‹+ â€‹MSCs group in vivo experiment (11.80 â€‹± â€‹1.11 vs 7.00 â€‹± â€‹1.76, P â€‹< â€‹0.01). The dorsal AF defect in Fib-T-G â€‹+ â€‹MSCs group (0.02 â€‹± â€‹0.01 â€‹mm2) was significantly smaller than that (0.13 â€‹± â€‹0.05 â€‹mm2) in Fib â€‹+ â€‹MSCs group (P â€‹< â€‹0.05). Immunohistochemical staining showed more positive staining of COL2A1 and Aggrecan in Fib-T-G â€‹+ â€‹MSCs group than in Fib â€‹+ â€‹MSCs group. Conclusion: Genipin crosslinked hydrogel can bond the interface of AF lesions and transfer strain force. Stress stimulation maintained by adhesive hydrogel promotes AF healing. The translational potential of this article: We believe the effect of stress stimulation could be concluded through this study and provides more ideals in mechanical effects for further research, which is a key technique for repairing intervertebral disc in clinic. The adhesive hydrogel of Fib-T-G+MSCs has low toxicity and helps bond the interface of AF lesion and transfer strain force, having great potential in the repair of AF lesion.

15.
Int J Mol Sci ; 24(12)2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37373396

RESUMEN

The spinal cord has a poor ability to regenerate after an injury, which may be due to cell loss, cyst formation, inflammation, and scarring. A promising approach to treating a spinal cord injury (SCI) is the use of biomaterials. We have developed a novel hydrogel scaffold fabricated from oligo(poly(ethylene glycol) fumarate) (OPF) as a 0.08 mm thick sheet containing polymer ridges and a cell-attractive surface on the other side. When the cells are cultured on OPF via chemical patterning, the cells attach, align, and deposit ECM along the direction of the pattern. Animals implanted with the rolled scaffold sheets had greater hindlimb recovery compared to that of the multichannel scaffold control, which is likely due to the greater number of axons growing across it. The immune cell number (microglia or hemopoietic cells: 50-120 cells/mm2 in all conditions), scarring (5-10% in all conditions), and ECM deposits (Laminin or Fibronectin: approximately 10-20% in all conditions) were equal in all conditions. Overall, the results suggest that the scaffold sheets promote axon outgrowth that can be guided across the scaffold, thereby promoting hindlimb recovery. This study provides a hydrogel scaffold construct that can be used in vitro for cell characterization or in vivo for future neuroprosthetics, devices, or cell and ECM delivery.


Asunto(s)
Organofosfonatos , Traumatismos de la Médula Espinal , Ratas , Animales , Hidrogeles/química , Organofosfonatos/metabolismo , Cicatriz/patología , Ratas Sprague-Dawley , Regeneración Nerviosa , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/metabolismo , Axones/patología , Andamios del Tejido/química
16.
Int J Stem Cells ; 16(3): 251-259, 2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37385634

RESUMEN

Mesenchymal stromal cells (MSCs) have attracted scientific and medical interest due to their self-renewing properties, pluripotency, and paracrine function. However, one of the main limitations to the clinical application of MSCs is their loss of efficacy after transplantation in vivo. Various bioengineering technologies to provide stem cell niche-like conditions have the potential to overcome this limitation. Here, focusing on the stem cell niche microenvironment, studies to maximize the immunomodulatory potential of MSCs by controlling biomechanical stimuli, including shear stress, hydrostatic pressure, stretch, and biophysical cues, such as extracellular matrix mimetic substrates, are discussed. The application of biomechanical forces or biophysical cues to the stem cell microenvironment will be beneficial for enhancing the immunomodulatory function of MSCs during cultivation and overcoming the current limitations of MSC therapy.

17.
Stem Cell Res Ther ; 14(1): 120, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37143116

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs), commonly sourced from adipose tissue, bone marrow and umbilical cord, have been widely used in many medical conditions due to their therapeutic potential. Yet, the still limited understanding of the underlying mechanisms of action hampers clinical translation. Clinical potency can vary considerably depending on tissue source, donor attributes, but importantly, also culture conditions. Lack of standard procedures hinders inter-study comparability and delays the progression of the field. The aim of this study was A- to assess the impact on MSC characteristics when different laboratories, performed analysis on the same MSC material using harmonised culture conditions and B- to understand source-specific differences. METHODS: Three independent institutions performed a head-to-head comparison of human-derived adipose (A-), bone marrow (BM-), and umbilical cord (UC-) MSCs using harmonised culture conditions. In each centre, cells from one specific tissue source were isolated and later distributed across the network to assess their biological properties, including cell expansion, immune phenotype, and tri-lineage differentiation (part A). To assess tissue-specific function, angiogenic and immunomodulatory properties and the in vivo biodistribution were compared in one expert lab (part B). RESULTS: By implementing a harmonised manufacturing workflow, we obtained largely reproducible results across three independent laboratories in part A of our study. Unique growth patterns and differentiation potential were observed for each tissue source, with similar trends observed between centres. Immune phenotyping verified expression of typical MSC surface markers and absence of contaminating surface markers. Depending on the established protocols in the different laboratories, quantitative data varied slightly. Functional experiments in part B concluded that conditioned media from BM-MSCs significantly enhanced tubulogenesis and endothelial migration in vitro. In contrast, immunomodulatory studies reported superior immunosuppressive abilities for A-MSCs. Biodistribution studies in healthy mice showed lung entrapment after administration of all three types of MSCs, with a significantly faster clearance of BM-MSCs. CONCLUSION: These results show the heterogeneous behaviour and regenerative properties of MSCs as a reflection of intrinsic tissue-origin properties while providing evidence that the use of harmonised culture procedures can reduce but do not eliminate inter-lab and operator differences.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Mesenquimatosas , Humanos , Animales , Ratones , Células Cultivadas , Distribución Tisular , Diferenciación Celular , Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/metabolismo , Proliferación Celular , Células de la Médula Ósea , Cordón Umbilical
18.
Int J Mol Sci ; 24(9)2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37175936

RESUMEN

The effects of the administration of mesenchymal stromal cells (MSC) may vary according to the source. We hypothesized that MSC-derived extracellular vesicles (EVs) obtained from bone marrow (BM), adipose (AD), or lung (L) tissues may also lead to different effects in sepsis. We profiled the proteome from EVs as a first step toward understanding their mechanisms of action. Polymicrobial sepsis was induced in C57BL/6 mice by cecal ligation and puncture (SEPSIS) and SHAM (control) animals only underwent laparotomy. Twenty-four hours after surgery, animals in the SEPSIS group were randomized to receive saline or 3 × 106 MSC-derived EVs from BM, AD, or L. The diffuse alveolar damage was decreased with EVs from all three sources. In kidneys, BM-, AD-, and L-EVs reduced edema and expression of interleukin-18. Kidney injury molecule-1 expression decreased only in BM- and L-EVs groups. In the liver, only BM-EVs reduced congestion and cell infiltration. The size and number of EVs from different sources were not different, but the proteome of the EVs differed. BM-EVs were enriched for anti-inflammatory proteins compared with AD-EVs and L-EVs. In conclusion, BM-EVs were associated with less organ damage compared with the other sources of EVs, which may be related to differences detected in their proteome.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Sepsis , Animales , Ratones , Vesículas Extracelulares/metabolismo , Pulmón , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Proteoma/metabolismo , Sepsis/metabolismo
19.
Biomedicines ; 11(5)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37239000

RESUMEN

Systemic sclerosis (SSc) is an immune-mediated disease wherein T cells are particularly implicated, presenting a poor prognosis and limited therapeutic options. Thus, mesenchymal-stem/stromal-cell (MSC)-based therapies can be of great benefit to SSc patients given their immunomodulatory, anti-fibrotic, and pro-angiogenic potential, which is associated with low toxicity. In this study, peripheral blood mononuclear cells from healthy individuals (HC, n = 6) and SSc patients (n = 9) were co-cultured with MSCs in order to assess how MSCs affected the activation and polarization of 58 different T cell subsets, including Th1, Th17, and Treg. It was found that MSCs downregulated the activation of 26 out of the 41 T cell subsets identified within CD4+, CD8+, CD4+CD8+, CD4-CD8-, and γδ T cells in SSc patients (HC: 29/42) and affected the polarization of 13 out of 58 T cell subsets in SSc patients (HC: 22/64). Interestingly, SSc patients displayed some T cell subsets with an increased activation status and MSCs were able to downregulate all of them. This study provides a wide-ranging perspective of how MSCs affect T cells, including minor subsets. The ability to inhibit the activation and modulate the polarization of several T cell subsets, including those implicated in SSc's pathogenesis, further supports the potential of MSC-based therapies to regulate T cells in a disease whose onset/development may be due to immune system's malfunction.

20.
Stem Cells Dev ; 32(13-14): 365-378, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37051687

RESUMEN

Articular cartilage injury is common in various conditions, including osteoarthritis, rheumatic diseases, and trauma. Current treatments for cartilage injury fail to completely regenerate the damaged cartilage. Mesenchymal stromal cells (MSCs) have emerged as potential candidates for cartilage regeneration. However, MSCs exhibit hypertrophic differentiation, and their chondrogenic ability is reduced in an inflammatory environment. In recent years, genetic modification has been proposed for optimizing MSC-based therapies, some of which are expected to enter clinical trials. This review summarizes recent research findings and developments in genetic engineering strategies to enhance stem cell-based therapy for cartilage regeneration. We also discuss the mechanisms of biofunctions of MSCs in cartilage regeneration and outline the efficacy and safety of the different genetic modification strategies, including viral and nonviral delivery transduction. Finally, we highlight the major challenges and prospects for clinical translation of genetically modified MSCs.


Asunto(s)
Cartílago Articular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Osteoartritis , Humanos , Diferenciación Celular/genética , Condrogénesis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA