Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.362
Filtrer
1.
FASEB J ; 38(14): e23798, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-38989582

RÉSUMÉ

The role of mesenchymal-stem-cell-derived exosomes (MSCs-Exo) in the regulation of macrophage polarization has been recognized in several diseases. There is emerging evidence that MSCs-Exo partially prevent the progression of diabetic nephropathy (DN). This study aimed to investigate whether exosomes secreted by MSCs pre-treated with a diabetic environment (Exo-pre) have a more pronounced protective effect against DN by regulating the balance of macrophages. Exo-pre and Exo-Con were isolated from the culture medium of UC-MSCs pre-treated with a diabetic mimic environment and natural UC-MSCs, respectively. Exo-pre and Exo-Con were injected into the tail veins of db/db mice three times a week for 6 weeks. Serum creatinine and serum urea nitrogen levels, the urinary protein/creatinine ratio, and histological staining were used to determine renal function and morphology. Macrophage phenotypes were analyzed by immunofluorescence, western blotting, and quantitative reverse transcription polymerase chain reaction. In vitro, lipopolysaccharide-induced M1 macrophages were incubated separately with Exo-Con and Exo-pre. We performed microRNA (miRNA) sequencing to identify candidate miRNAs and predict their target genes. An miRNA inhibitor was used to confirm the role of miRNAs in macrophage modulation. Exo-pre were more potent than Exo-Con at alleviating DN. Exo-pre administration significantly reduced the number of M1 macrophages and increased the number of M2 macrophages in the kidney compared to Exo-Con administration. Parallel outcomes were observed in the co-culture experiments. Moreover, miR-486-5p was distinctly expressed in Exo-Con and Exo-pre groups, and it played an important role in macrophage polarization by targeting PIK3R1 through the PI3K/Akt pathway. Reducing miR-486-5p levels in Exo-pre abolished macrophage polarization modulation. Exo-pre administration exhibited a superior effect on DN by remodeling the macrophage balance by shuttling miR-486-5p, which targets PIK3R1.


Sujet(s)
Néphropathies diabétiques , Exosomes , Macrophages , Cellules souches mésenchymateuses , microARN , Cordon ombilical , Exosomes/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Néphropathies diabétiques/métabolisme , Souris , Macrophages/métabolisme , microARN/métabolisme , microARN/génétique , Cordon ombilical/cytologie , Cordon ombilical/métabolisme , Mâle , Souris de lignée C57BL , Activation des macrophages
2.
J Biomed Sci ; 31(1): 71, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39004727

RÉSUMÉ

BACKGROUND: Critical limb-threatening ischemia (CLTI) constitutes the most severe manifestation of peripheral artery disease, usually induced by atherosclerosis. CLTI patients suffer from high risk of amputation of the lower extremities and elevated mortality rates, while they have low options for surgical revascularization due to associated comorbidities. Alternatively, cell-based therapeutic strategies represent an effective and safe approach to promote revascularization. However, the variability seen in several factors such as cell combinations or doses applied, have limited their success in clinical trials, being necessary to reach a consensus regarding the optimal "cellular-cocktail" prior further application into the clinic. To achieve so, it is essential to understand the mechanisms by which these cells exert their regenerative properties. Herein, we have evaluated, for the first time, the regenerative and vasculogenic potential of a combination of endothelial colony forming cells (ECFCs) and mesenchymal stem cells (MSCs) isolated from adipose-tissue (AT), compared with ECFCs from umbilical cord blood (CB-ECFCs) and AT-MSCs, in a murine model of CLTI. METHODS: Balb-c nude mice (n:32) were distributed in four different groups (n:8/group): control shams, and ischemic mice (after femoral ligation) that received 50 µl of physiological serum alone or a cellular combination of AT-MSCs with either CB-ECFCs or AT-ECFCs. Follow-up of blood flow reperfusion and ischemic symptoms was carried out for 21 days, when mice were sacrificed to evaluate vascular density formation. Moreover, the long-term molecular changes in response to CLTI and both cell combinations were analyzed in a proteomic quantitative approach. RESULTS: AT-MSCs with either AT- or CB-ECFCs, promoted a significant recovery of blood flow in CLTI mice 21 days post-ischemia. Besides, they modulated the inflammatory and necrotic related processes, although the CB group presented the slowest ischemic progression along the assay. Moreover, many proteins involved in the repairing mechanisms promoted by cell treatments were identified. CONCLUSIONS: The combination of AT-MSCs with AT-ECFCs or with CB-ECFCs promoted similar revascularization in CLTI mice, by restoring blood flow levels, together with the modulation of the inflammatory and necrotic processes, and reduction of muscle damage. The protein changes identified are representative of the molecular mechanisms involved in ECFCs and MSCs-induced revascularization (immune response, vascular repair, muscle regeneration, etc.).


Sujet(s)
Tissu adipeux , Modèles animaux de maladie humaine , Ischémie , Cellules souches mésenchymateuses , Souris de lignée BALB C , Souris nude , Animaux , Souris , Ischémie/thérapie , Ischémie/physiopathologie , Cordon ombilical/cytologie , Mâle , Transplantation de cellules souches mésenchymateuses/méthodes , Néovascularisation physiologique , Cellules endothéliales , Humains
3.
J Vis Exp ; (209)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39037235

RÉSUMÉ

Mesenchymal stem cells (MSCs) are a population of multipotent cells with remarkable regenerative and immunomodulatory properties. Wharton's jelly (WJ) from the umbilical cord (UC) has gained increasing interest in the biomedical field as an outstanding source of MSCs. However, challenges such as limited supply and lack of standardization in existing methods have arisen. This article presents a novel method for enhancing MSC yield by dissecting intact WJ from the umbilical cord. The method employs blunt dissection to remove the epithelial layer, maintaining the integrity of the entire WJ and resulting in an increased quantity and viability of harvested MSCs. This approach significantly reduces WJ waste compared to conventional sharp dissection methods. To ensure the purity of WJ-MSCs and minimize external cellular influence, a procedure utilizing internal tension to peel off the endothelium after flipping the UC was conducted. Additionally, the Petri dish was inverted for a short time during explant culture to improve attachment and cell outgrowth. Comparative analysis demonstrated the superiority of the proposed method, showing a higher yield of WJ and WJ-MSCs with better viability than traditional methods. The similar morphology and expression pattern of cell surface markers in both methods confirm their characterization and purity for various applications. This method provides a high-yield and high-viability approach for WJ-MSC isolation, demonstrating great potential for the clinical application of MSCs.


Sujet(s)
Cellules souches mésenchymateuses , Cordon ombilical , Cellules souches mésenchymateuses/cytologie , Cordon ombilical/cytologie , Humains , Gelée de Wharton/cytologie , Séparation cellulaire/méthodes , Techniques cytologiques/méthodes , Femelle
4.
Cell Mol Life Sci ; 81(1): 297, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992309

RÉSUMÉ

Muse cells, identified as cells positive for the pluripotent surface marker SSEA-3, are pluripotent-like endogenous stem cells located in the bone marrow (BM), peripheral blood, and organ connective tissues. The detailed characteristics of SSEA-3(+) cells in extraembryonic tissue, however, are unknown. Here, we demonstrated that similar to human-adult tissue-Muse cells collected from the BM, adipose tissue, and dermis as SSEA-3(+), human-umbilical cord (UC)-SSEA-3(+) cells express pluripotency markers, differentiate into triploblastic-lineage cells at a single cell level, migrate to damaged tissue, and exhibit low telomerase activity and non-tumorigenicity. Notably, ~ 20% of human-UC-SSEA-3(+) cells were negative for X-inactive specific transcript (XIST), a naïve pluripotent stem cell characteristic, whereas all human adult tissue-Muse cells are XIST-positive. Single-cell RNA sequencing revealed that the gene expression profile of human-UC-SSEA-3(+) cells was more similar to that of human post-implantation blastocysts than human-adult tissue-Muse cells. The DNA methylation level showed the same trend, and notably, the methylation levels in genes particularly related to differentiation were lower in human-UC-SSEA-3(+) cells than in human-adult tissue-Muse cells. Furthermore, human-UC-SSEA-3(+) cells newly express markers specific to extraembryonic-, germline-, and hematopoietic-lineages after differentiation induction in vitro whereas human-adult tissue-Muse cells respond only partially to the induction. Among various stem/progenitor cells in living bodies, those that exhibit properties similar to post-implantation blastocysts in a naïve state have not yet been found in humans. Easily accessible human-UC-SSEA-3(+) cells may be a valuable tool for studying early-stage human development and human reproductive medicine.


Sujet(s)
Blastocyste , Différenciation cellulaire , Antigènes embryonnaires spécifiques de stade , Cordon ombilical , Humains , Antigènes embryonnaires spécifiques de stade/métabolisme , Cordon ombilical/cytologie , Blastocyste/cytologie , Blastocyste/métabolisme , Antigènes glycanniques associés aux tumeurs/métabolisme , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , Analyse sur cellule unique , Telomerase/métabolisme , Telomerase/génétique , Femelle
5.
Stem Cell Res Ther ; 15(1): 210, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39020429

RÉSUMÉ

BACKGROUND: Hemophilia B is an X-linked bleeding disorder caused by a mutation in the gene responsible for encoding coagulation factor IX (FIX). Gene therapy offers promising potential for curing this disease. However, the current method of relatively high dosage of virus injection carries inherent risks. The purpose of this study was to introduce a novel scAAV-DJ/8-LP1-hFIXco vector transduced human umbilical cord blood derived mesenchymal stem cells (HUCMSCs) as an alternative cell-based gene therapy to conventional gene therapy for Hemophilia B. METHODS: The LP1-hFIXco gene structure was designed by us through searching the literature from NCBI and the scAAV-DJ/8-LP1-hFIXco vector was constructed by a commercial company. The HUCMSCs were cultivated in routine approach and transduced with scAAV-DJ/8-LP1-hFIXco vector. The human FIX activation system was employed for detection of hFIXco activity. The RNA and protein expression levels of the hFIXco were evaluated using PCR and western blot techniques. In animal studies, both NSG and F9-KO mice were used for the experiment, in which clotting time was utilized as a parameter for bleeding assessment. The immunohistochemical analysis was used to assess the distribution of HUCMSCs in mouse tissue sections. The safety for tumorigenicity of this cell-based gene therapy was evaluated by pathological observation after hematoxylin-eosin staining. RESULTS: The transduction of HUCMSCs with the scAAV-DJ/8-LP1-hFIXco vector results in consistent and sustainable secretion of human FIXco during 5 months period both in vitro and in mouse model. The secretion level (hFIXco activity: 97.1 ± 2.3% at day 7 to 48.8 ± 4.5% at 5 months) was comparable to that observed following intravenous injection with a high dose of the viral vector (hFIXco activity: 95.2 ± 2.2% to 40.8 ± 4.3%). After a 5-month observation period, no clonal expansions of the transduced cells in tissues were observed in any of the mice studied. CONCLUSIONS: We have discovered a novel and safer HUCMSCs mediated approach potentially effective for gene therapy in hemophilia B.


Sujet(s)
Facteur IX , Thérapie génétique , Vecteurs génétiques , Hémophilie B , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Humains , Thérapie génétique/méthodes , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Hémophilie B/thérapie , Hémophilie B/génétique , Souris , Facteur IX/génétique , Facteur IX/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Vecteurs génétiques/génétique , Vecteurs génétiques/métabolisme , Transduction génétique , Cordon ombilical/cytologie , Souris knockout , Sang foetal/cytologie , Sang foetal/métabolisme
6.
Stem Cell Res Ther ; 15(1): 216, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39020448

RÉSUMÉ

BACKGROUND: Human umbilical cord mesenchymal stem cells-derived extracellular vesicles (hUCMSC-EVs) have potent immunomodulatory properties similar to parent cells. This study investigated the therapeutic effects and immunomodulatory mechanisms of hUCMSC-EVs in an experimental lupus nephritis model. METHODS: The hUCMSC-EVs were isolated by using differential ultracentrifugation. In vivo, the therapeutic effects of hUCMSC-EVs in lupus-prone MRL/lpr mice were investigated, and the mechanisms of treatment were explored according to the abnormal T and B cell responses among both the spleen and kidney. RESULTS: MRL/lpr mice treated with hUCMSC-EVs reduced proteinuria extent, serum creatinine and renal pathological damage; decreased splenic index and serum anti-dsDNA IgG level; and improved survival rate. hUCMSC-EVs lowered the percentage of T helper (Th)1 cells, double-negative T (DNT) cells, and plasma cells among splenocytes; inhibited the infiltration of Th17 cells but promoted regulatory T (Treg) cells in the kidney, followed by a reduction in pro-inflammatory cytokine levels(IFN-γ, IL-2, IL-6, IL-21, and IL-17 A). In addition, hUCMSC-EVs inhibited the activation of STAT3 and down-regulated IL-17 A protein levels in the kidney. CONCLUSION: The results of this study demonstrated that hUCMSC-EVs had therapeutic effects on experimental lupus nephritis (LN) by regulating Th1/Th17/Treg imbalance and inhibiting DNT and plasma cells. Additionally, hUCMSC-EVs inhibited Th17 cell differentiation in kidney by regulating the IL-6/STAT3/IL-17 signal pathway, which might be an important mechanism for alleviating renal injury. Taken together, we demonstrated that hUCMSC-EVs regulating T and B cell immune responses might represent a novel mechanism of hUCMSCs in treating LN, thus providing a new strategy for treating LN.


Sujet(s)
Vésicules extracellulaires , Glomérulonéphrite lupique , Cellules souches mésenchymateuses , Souris de lignée MRL lpr , Glomérulonéphrite lupique/thérapie , Glomérulonéphrite lupique/immunologie , Glomérulonéphrite lupique/anatomopathologie , Glomérulonéphrite lupique/métabolisme , Animaux , Vésicules extracellulaires/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Souris , Humains , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Femelle , Facteur de transcription STAT-3/métabolisme , Cellules Th17/immunologie , Cellules Th17/métabolisme , Rate , Modèles animaux de maladie humaine , Rein/anatomopathologie , Rein/métabolisme , Cytokines/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Cordon ombilical/cytologie
7.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 681-689, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38949136

RÉSUMÉ

Objective: To investigate the therapeutic effect of methotrexate loaded vesicles on experimental periodontitis in mice. Methods: Extracellular vesicles (EVs) were isolated from human umbilical cord mesenchymal stem cells (hUC-MSC). Methotrexate loaded vesicles (MTX-EVs) were constructed, whose morphology and size were analyzed by using scanning electron microscopy and particle size analyzer. Western blotting was used to identify their surface specific proteins. C57BL/6J male mice of 4-5 weeks (provided by Experimental Animal Center of The Fourth Military Medical University) were selected, among which 8 were randomly selected by blind grasp method without treatment and fed normally as normal group, and others were induced to periodontitis models by local injection of lipopolysaccharide (LPS) into the periodontium. The LPS was injected once every day with a concentration of 2 g/L and a volume of 5 µl, lasting for two weeks. The mice with successfully induced periodontitis were randomly divided into 4 groups by blind grasping method, with 8 mice in each group. The LPS group was with no treatment, and the other three groups were treated with periodontal local injection of MTX, EVs or MTX-EVs, respectively. Two weeks later, enzyme-linked immunosorbent assay (ELISA) was used to detect the expressions of inflammatory cytokine interleukin (IL)-1ß, IL-6 and tumor necrosis factor-α (TNF-α) in gingival tissue. The amount of alveolar bone resorption of four groups was detected by using micro-CT scanning and HE staining. The expression proportion of the inflammatory factor in gingival tissue was analyzed by using flow cytometry. Results: The scanning electron microscopy results showed that EVs and MTX-EVs were circular or elliptical in shape. Dynamic light scattering (DLS) particle size analysis showed that the particle size of EVs was around 200 nm, while that of MTX-EVs was around 300 nm. The ELISA results showed IL-1ß levels in the normal group, LPS group, LPS+MTX group, LPS+EVs group and LPS+MTX-EVs group were (28.86±2.76), (51.50±2.04), (35.26±2.40), (45.49±2.04) and (35.77±3.49) ng/L. That is, the IL-1ß concentrations in the LPS+MTX group, LPS+EVs group and LPS+MTX-EVs group were significantly lower than that in the LPS group (P<0.05); the mass concentration of IL-1ß in the LPS +MTX-EVs group was significantly lower than that in the LPS+EVs group (P<0.05). The concentrations of IL-6 in the normal group, LPS group, LPS+MTX group, LPS+EVs group and LPS+MTX-EVs group were (125.44±4.12), (221.64±10.59), (178.16±16.90), (181.09±18.22) and (170.15±9.04) ng/L, among which the concentration of IL-6 in the last three groups were significantly lower than that in the LPS group (P<0.05). The mass concentration of IL-6 in the LPS+MTX-EVs group was significantly lower than those in the LPS+MTX group and LPS+EVs group (P<0.05). The concentrations of TNF-α in the normal group, LPS group, LPS+MTX group, LPS+EVs group and LPS+MTX-EVs group were (320.27±38.68), (479.62±40.94), (342.18±25.89), (415.88±12.01) and (325.75±30.83) ng/L, among which the concentrations of last three groups were significantly lower than the LPS group (P<0.05); the mass concentration of TNF-α in the LPS+MTX-EVs group was significantly lower than those in the LPS+EVs group and LPS+MTX group (P<0.05). The micro-CT results showed that the distance of cement-enamel junction-alveolar bone crest (CEJ-ABC) of the first molar and root (M1R1) in the normal group, LPS group, LPS+MTX group, LPS+EVs group and LPS+MTX-EVs group of mice were (0.11±0.03), (0.28±0.02), (0.23±0.03), (0.20±0.04), and (0.18±0.03) mm, respectively. Compared with the LPS group, the CEJ-ABC of the M1R1 in the LPS+MTX group, LPS+EVs group and LPS+MTX-EVs group were inhibited to varied degrees with statistically significant differences (P<0.05). Among them, LPS+MTX-EVs group had the best bone resorption inhibitioin effect compared to LPS+MTX group and LPS+EVs group, and the differences were statistically significant (P<0.05). The flow cytometry results indicated that the proportion of interferon-γ (IFN-γ) positive cells was (11.77±1.02)% in the LPS group, (6.87±0.65)% in the LPS+EVs group, and (4.15±0.92)% in the LPS+MTX-EVs group, respectively. The proportions of IFN-γ positive cells in the LPS+EVs group and LPS+MTX-EVs group were significantly lower than that in the LPS group (P<0.05), while the ratio of IFN-γ positive cells in the LPS+MTX-EVs group was found significantly lower than that in the LPS+EVs group (P<0.05). Conclusions: MTX-EVs can effectively alleviate the periodontal local inflammatory environment and reduce bone resorption of alveolar bone in periodontitis model mice.


Sujet(s)
Vésicules extracellulaires , Interleukine-1 bêta , Interleukine-6 , Lipopolysaccharides , Méthotrexate , Souris de lignée C57BL , Parodontite , Facteur de nécrose tumorale alpha , Animaux , Parodontite/thérapie , Parodontite/traitement médicamenteux , Vésicules extracellulaires/métabolisme , Souris , Mâle , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6/métabolisme , Interleukine-1 bêta/métabolisme , Cellules souches mésenchymateuses/cytologie , Modèles animaux de maladie humaine , Humains , Cordon ombilical/cytologie , Interféron gamma/métabolisme
8.
Stem Cell Res Ther ; 15(1): 196, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956734

RÉSUMÉ

Over the past decade, we have witnessed the development of cell transplantation as a new strategy for repairing spinal cord injury (SCI). However, due to the complexity of the central nervous system (CNS), achieving successful clinical translation remains a significant challenge. Human umbilical cord mesenchymal stem cells (hUMSCs) possess distinct advantages, such as easy collection, lack of ethical concerns, high self-renewal ability, multilineage differentiation potential, and immunomodulatory properties. hUMSCs are promising for regenerating the injured spinal cord to a significant extent. At the same time, for advancing SCI treatment, the appropriate benefit and risk evaluation methods play a pivotal role in determining the clinical applicability of treatment plans. Hence, this study discusses the advantages and risks of hUMSCs in SCI treatment across four dimensions-comprehensive evaluation of motor and sensory function, imaging, electrophysiology, and autonomic nervous system (ANS) function-aiming to improve the rationality of relevant clinical research and the feasibility of clinical translation.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Traumatismes de la moelle épinière , Cordon ombilical , Humains , Traumatismes de la moelle épinière/thérapie , Transplantation de cellules souches mésenchymateuses/méthodes , Cordon ombilical/cytologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Appréciation des risques , Différenciation cellulaire , Animaux
9.
Sci Rep ; 14(1): 15113, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956421

RÉSUMÉ

The aims of this study were to determine whether human umbilical cord mesenchymal stem cells (hucMSCs) modified by miRNA-25-3p (miR-25-3p) overexpression could promote venous endothelial cell proliferation and attenuate portal endothelial cell injury. HucMSCs and human umbilical vein endothelial cells (HUVEC) were isolated and cultured from human umbilical cord and characterized. Lentiviral vectors expressing miRNA-25-3p were transfected into hucMSCs and confirmed by PCR. We verified the effect of miR-25-3p-modified hucMSCs on HUVEC by cell co-culture and cell supernatant experiments. Subsequently, exosomes of miR-25-3p-modified hucMSCs were isolated from cell culture supernatants and characterized by WB, NTA and TEM. We verified the effects of miR-25-3p-modified exosomes derived from hucMSCs on HUVEC proliferation, migration, and angiogenesis by in vitro cellular function experiments. Meanwhile, we further examined the downstream target genes and signaling pathways potentially affected by miR-25-3p-modified hucMSC-derived exosomes in HUVEC. Finally, we established a rat portal vein venous thrombosis model by injecting CM-DiR-labeled hucMSCs intravenously into rats and examining the homing of cells in the portal vein by fluorescence microscopy. Histological and immunohistochemical experiments were used to examine the effects of miRNA-25-3p-modified hucMSCs on the proliferation and damage of portal vein endothelial cells. Primary hucMSCs and HUVECs were successfully isolated, cultured and characterized. Primary hucMSCs were modified with a lentiviral vector carrying miR-25-3p at MOI 80. Co-culture and cell supernatant intervention experiments showed that overexpression of miRNA-25-3p in hucMSCs enhanced HUVEC proliferation, migration and tube formation in vitro. We successfully isolated and characterized exosomes of miR-25-3p-modified hucMSCs, and exosome intervention experiments demonstrated that miR-25-3p-modified exosomes derived from hucMSCs similarly enhanced the proliferation, migration, and angiogenesis of HUVECs. Subsequent PCR and WB analyses indicated PTEN/KLF4/AKT/ERK1/2 as potential pathways of action. Analysis in a rat portal vein thrombosis model showed that miR-25-3p-modified hucMSCs could homing to damaged portal veins. Subsequent histological and immunohistochemical examinations demonstrated that intervention with miR-25-3p overexpression-modified hucMSCs significantly reduced damage and attenuated thrombosis in rat portal veins. The above findings indicate suggest that hucMSCs based on miR-25-3p modification may be a promising therapeutic approach for use in venous thrombotic diseases.


Sujet(s)
Prolifération cellulaire , Exosomes , Cellules endothéliales de la veine ombilicale humaine , Cellules souches mésenchymateuses , microARN , Veine porte , microARN/génétique , microARN/métabolisme , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Rats , Exosomes/métabolisme , Exosomes/génétique , Veine porte/métabolisme , Mouvement cellulaire/génétique , Rat Sprague-Dawley , Mâle , Thrombose veineuse/génétique , Thrombose veineuse/métabolisme , Thrombose veineuse/anatomopathologie , Thrombose veineuse/thérapie , Cellules cultivées , Techniques de coculture , Transduction du signal , Cordon ombilical/cytologie
10.
Stem Cell Res Ther ; 15(1): 190, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956621

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) demonstrate a wide range of therapeutic capabilities in the treatment of inflammatory bowel disease (IBD). The intraperitoneal injection of MSCs has exhibited superior therapeutic efficacy on IBD than intravenous injection. Nevertheless, the precise in vivo distribution of MSCs and their biological consequences following intraperitoneal injection remain inadequately understood. Additional studies are required to explore the correlation between MSCs distribution and their biological effects. METHODS: First, the distribution of human umbilical cord MSCs (hUC-MSCs) and the numbers of Treg and Th17 cells in mesenteric lymph nodes (MLNs) were analyzed after intraperitoneal injection of hUC-MSCs. Subsequently, the investigation focused on the levels of transforming growth factor beta1 (TGF-ß1), a key cytokine to the biology of both Treg and Th17 cells, in tissues of mice with colitis, particularly in MLNs. The study also delved into the impact of hUC-MSCs therapy on Treg cell counts in MLNs, as well as the consequence of TGFB1 knockdown hUC-MSCs on the differentiation of Treg cells and the treatment of IBD. RESULTS: The therapeutic effectiveness of intraperitoneally administered hUC-MSCs in the treatment of colitis was found to be significant, which was closely related to their quick migration to MLNs and secretion of TGF-ß1. The abundance of hUC-MSCs in MLNs of colitis mice is much higher than that in other organs even the inflamed sites of colon. Intraperitoneal injection of hUC-MSCs led to a significant increase in the number of Treg cells and a decrease in Th17 cells especially in MLNs. Furthermore, the concentration of TGF-ß1, the key cytokine for Treg differentiation, were also found to be significantly elevated in MLNs after hUC-MSCs treatment. Knockdown of TGFB1 in hUC-MSCs resulted in a noticeable reduction of Treg cells in MLNs and the eventually failure of hUC-MSCs therapy in colitis. CONCLUSIONS: MLNs may be a critical site for the regulatory effect of hUC-MSCs on Treg/Th17 cells and the therapeutic effect on colitis. TGF-ß1 derived from hUC-MSCs promotes local Treg differentiation in MLNs. This study will provide new ideas for the development of MSC-based therapeutic strategies in IBD patients.


Sujet(s)
Différenciation cellulaire , Colite , Noeuds lymphatiques , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Lymphocytes T régulateurs , Cellules Th17 , Facteur de croissance transformant bêta-1 , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Humains , Colite/thérapie , Colite/induit chimiquement , Colite/métabolisme , Colite/anatomopathologie , Transplantation de cellules souches mésenchymateuses/méthodes , Souris , Noeuds lymphatiques/métabolisme , Cellules Th17/métabolisme , Cellules Th17/immunologie , Cordon ombilical/cytologie , Mésentère/métabolisme , Souris de lignée C57BL , Souris de lignée BALB C , Mâle , Maladies inflammatoires intestinales/thérapie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie
11.
PeerJ ; 12: e17616, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952966

RÉSUMÉ

Background: Mesenchymal stem cells (MSCs) are increasingly recognized for their regenerative potential. However, their clinical application is hindered by their inherent variability, which is influenced by various factors, such as the tissue source, culture conditions, and passage number. Methods: MSCs were sourced from clinically relevant tissues, including adipose tissue-derived MSCs (ADMSCs, n = 2), chorionic villi-derived MSCs (CMMSCs, n = 2), amniotic membrane-derived MSCs (AMMSCs, n = 3), and umbilical cord-derived MSCs (UCMSCs, n = 3). Passages included the umbilical cord at P0 (UCMSCP0, n = 2), P3 (UCMSCP3, n = 2), and P5 (UCMSCP5, n = 2) as well as the umbilical cord at P5 cultured under low-oxygen conditions (UCMSCP5L, n = 2). Results: We observed that MSCs from different tissue origins clustered into six distinct functional subpopulations, each with varying proportions. Notably, ADMSCs exhibited a higher proportion of subpopulations associated with vascular regeneration, suggesting that they are beneficial for applications in vascular regeneration. Additionally, CMMSCs had a high proportion of subpopulations associated with reproductive processes. UCMSCP5 and UCMSCP5L had higher proportions of subpopulations related to female reproductive function than those for earlier passages. Furthermore, UCMSCP5L, cultured under low-oxygen (hypoxic) conditions, had a high proportion of subpopulations associated with pro-angiogenic characteristics, with implications for optimizing vascular regeneration. Conclusions: This study revealed variation in the distribution of MSC subpopulations among different tissue sources, passages, and culture conditions, including differences in functions related to vascular and reproductive system regeneration. These findings hold promise for personalized regenerative medicine and may lead to more effective clinical treatments across a spectrum of medical conditions.


Sujet(s)
Tissu adipeux , Cellules souches mésenchymateuses , Cordon ombilical , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/physiologie , Humains , Cordon ombilical/cytologie , Femelle , Tissu adipeux/cytologie , Cellules cultivées , Villosités choriales/physiologie , Amnios/cytologie , Différenciation cellulaire
12.
J Biomed Mater Res B Appl Biomater ; 112(7): e35448, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38968133

RÉSUMÉ

Traditional decellularized bioscaffolds possessing intact vascular networks and unique architecture have been extensively studied as conduits for repairing nerve damage. However, they are limited by the absence of electrical conductivity, which is crucial for proper functioning of nervous tissue. This study focuses on investigating decellularized umbilical cord arteries by applying coatings of graphene oxide (GO) and reduced graphene oxide (RGO) to their inner surfaces. This resulted in a homogeneous GO coating that fully covered the internal lumen of the arteries. The results of electrical measurements demonstrated that the conductivity of the scaffolds could be significantly enhanced by incorporating RGO and GO conductive sheets. At a low frequency of 0.1 Hz, the electrical resistance level of the coated scaffolds decreased by 99.8% with RGO and 98.21% with GO, compared with uncoated scaffolds. Additionally, the mechanical properties of the arteries improved by 24.69% with GO and 32.9% with RGO after the decellularization process. The GO and RGO coatings did not compromise the adhesion of endothelial cells and promoted cell growth. The cytotoxicity tests revealed that cell survival rate increased over time with RGO, while it decreased with GO, indicating the time-dependent effect on the cytotoxicity of GO and RGO. Blood compatibility evaluations showed that graphene nanomaterials did not induce hemolysis but exhibited some tendency toward blood coagulation.


Sujet(s)
Matériaux revêtus, biocompatibles , Conductivité électrique , Graphite , Artères ombilicales , Graphite/composition chimique , Humains , Matériaux revêtus, biocompatibles/composition chimique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Structures d'échafaudage tissulaires/composition chimique , Test de matériaux , Cordon ombilical/cytologie , Animaux
13.
Stem Cell Res Ther ; 15(1): 239, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080783

RÉSUMÉ

BACKGROUND: The leading cause of end-stage renal disease (ESRD) is diabetic nephropathy (DN). Podocyte damage is an early event in the development of DN. Currently, there is no effective treatment strategy that can slow the progression of DN or reverse its onset. The role of mesenchymal stem cells (MSCs) transplantation in diabetes and its complications has been extensively studied, and diabetic nephropathy has been a major focus. Irbesartan exerts reno-protective effects independent of lowering blood pressure, can reduce the incidence of proteinuria in rats, and is widely used clinically. However, it remains undetermined whether the combined utilization of the angiotensin II receptor antagonist irbesartan and MSCs could enhance efficacy in addressing DN. METHODS: A commonly used method for modeling type 2 diabetic nephropathy (T2DN) was established using a high-fat diet and a single low-dose injection of STZ (35 mg/kg). The animals were divided into the following 5 groups: (1) the control group (CON), (2) the diabetic nephropathy group (DN), (3) the mesenchymal stem cells treatment group (MSCs), (4) the irbesartan treatment group (Irb), and (5) the combined administration group (MSC + Irb). MSCs (2 × 106 cells/rat) were injected every 10 days through the tail vein for a total of three injections; irbesartan (30 mg/kg/d) was administered by gavage. Additionally, the safety and homing of mesenchymal stem cells were verified using positron emission tomography (PET) imaging. RESULTS: The combination treatment significantly reduced the UACR, kidney index, IGPTT, HOMA-IR, BUN, serum creatine, and related inflammatory factor levels and significantly improved renal function parameters and the expression of proteins related to glomerular podocyte injury in rats. Moreover, MSCs can homing target to damaged kidneys. CONCLUSIONS: Compared to the administration of MSCs or irbesartan alone, the combination of MSCs and irbesartan exerted better protective effects on glomerular podocyte injury, providing new ideas for the clinical application of mesenchymal stem cells.


Sujet(s)
Diabète expérimental , Néphropathies diabétiques , Irbésartan , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Podocytes , Animaux , Irbésartan/pharmacologie , Irbésartan/usage thérapeutique , Podocytes/effets des médicaments et des substances chimiques , Podocytes/anatomopathologie , Transplantation de cellules souches mésenchymateuses/méthodes , Rats , Cellules souches mésenchymateuses/métabolisme , Diabète expérimental/thérapie , Diabète expérimental/complications , Néphropathies diabétiques/thérapie , Néphropathies diabétiques/traitement médicamenteux , Mâle , Cordon ombilical/cytologie , Rat Sprague-Dawley , Humains , Transplantation hétérologue , Dérivés du biphényle/pharmacologie , Dérivés du biphényle/usage thérapeutique
14.
Stem Cell Res Ther ; 15(1): 229, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075621

RÉSUMÉ

BACKGROUND: Human mesenchymal stem cells originating from umbilical cord matrix are a promising therapeutic resource, and their differentiated cells are spotlighted as a tissue regeneration treatment. However, there are limitations to the medical use of differentiated cells from human umbilical cord matrix-mesenchymal stem cells (hUCM-MSCs), such as efficient differentiation methods. METHODS: To effectively differentiate hUCM-MSCs into hepatocyte-like cells (HLCs), we used the ROCK inhibitor, fasudil, which is known to induce endoderm formation, and gelatin, which provides extracellular matrix to the differentiated cells. To estimate a differentiation efficiency of early stage according to combination of gelatin and fasudil, transcription analysis was conducted. Moreover, to demonstrate that organelle states affect differentiation, we performed transcription, tomographic, and mitochondrial function analysis at each stage of hepatic differentiation. Finally, we evaluated hepatocyte function based on the expression of mRNA and protein, secretion of albumin, and activity of CYP3A4 in mature HLCs. RESULTS: Fasudil induced endoderm-related genes (GATA4, SOX17, and FOXA2) in hUCM-MSCs, and it also induced lipid droplets (LDs) inside the differentiated cells. However, the excessive induction of LDs caused by fasudil inhibited mitochondrial function and prevented differentiation into hepatoblasts. To prevent the excessive LDs formation, we used gelatin as a coating material. When hUCM-MSCs were induced into hepatoblasts with fasudil on high-viscosity (1%) gelatin-coated dishes, hepatoblast-related genes (AFP and HNF4A) showed significant upregulation on high-viscosity gelatin-coated dishes compared to those treated with low-viscosity (0.1%) gelatin. Moreover, other germline cell fates, such as ectoderm and mesoderm, were repressed under these conditions. In addition, LDs abundance was also reduced, whereas mitochondrial function was increased. On the other hand, unlike early stage of the differentiation, low viscosity gelatin was more effective in generating mature HLCs. In this condition, the accumulation of LDs was inhibited in the cells, and mitochondria were activated. Consequently, HLCs originated from hUCM-MSCs were genetically and functionally more matured in low-viscosity gelatin. CONCLUSIONS: This study demonstrated an effective method for differentiating hUCM-MSCs into hepatic cells using fasudil and gelatin of varying viscosities. Moreover, we suggest that efficient hepatic differentiation and the function of hepatic cells differentiated from hUCM-MSCs depend not only on genetic changes but also on the regulation of organelle states.


Sujet(s)
5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine , Différenciation cellulaire , Gélatine , Hépatocytes , Cellules souches mésenchymateuses , Cordon ombilical , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , 5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine/analogues et dérivés , 5-(2-Méthyl-pipérazine-1-sulfonyl)isoquinoléine/pharmacologie , Gélatine/composition chimique , Gélatine/pharmacologie , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/cytologie , Cordon ombilical/cytologie , Viscosité , Cellules cultivées , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques
15.
J Zhejiang Univ Sci B ; 25(7): 568-580, 2024 Jul 10.
Article de Anglais, Chinois | MEDLINE | ID: mdl-39011677

RÉSUMÉ

Diabetes mellitus (DM) is a disease syndrome characterized by chronic hyperglycaemia. A long-term high-glucose environment leads to reactive oxygen species (ROS) production and nuclear DNA damage. Human umbilical cord mesenchymal stem cell (HUcMSC) infusion induces significant antidiabetic effects in type 2 diabetes mellitus (T2DM) rats. Insulin-like growth factor 1 (IGF1) receptor (IGF1R) is important in promoting glucose metabolism in diabetes; however, the mechanism by which HUcMSC can treat diabetes through IGF1R and DNA damage repair remains unclear. In this study, a DM rat model was induced with high-fat diet feeding and streptozotocin (STZ) administration and rats were infused four times with HUcMSC. Blood glucose, interleukin-6 (IL-6), IL-10, glomerular basement membrane, and renal function were examined. Proteins that interacted with IGF1R were determined through coimmunoprecipitation assays. The expression of IGF1R, phosphorylated checkpoint kinase 2 (p-CHK2), and phosphorylated protein 53 (p-p53) was examined using immunohistochemistry (IHC) and western blot analysis. Enzyme-linked immunosorbent assay (ELISA) was used to determine the serum levels of 8-hydroxydeoxyguanosine (8-OHdG). Flow cytometry experiments were used to detect the surface markers of HUcMSC. The identification of the morphology and phenotype of HUcMSC was performed by way of oil red "O" staining and Alizarin red staining. DM rats exhibited abnormal blood glucose and IL-6/10 levels and renal function changes in the glomerular basement membrane, increased the expression of IGF1 and IGF1R. IGF1R interacted with CHK2, and the expression of p-CHK2 was significantly decreased in IGF1R-knockdown cells. When cisplatin was used to induce DNA damage, the expression of p-CHK2 was higher than that in the IGF1R-knockdown group without cisplatin treatment. HUcMSC infusion ameliorated abnormalities and preserved kidney structure and function in DM rats. The expression of IGF1, IGF1R, p-CHK2, and p-p53, and the level of 8-OHdG in the DM group increased significantly compared with those in the control group, and decreased after HUcMSC treatment. Our results suggested that IGF1R could interact with CHK2 and mediate DNA damage. HUcMSC infusion protected against kidney injury in DM rats. The underlying mechanisms may include HUcMSC-mediated enhancement of diabetes treatment via the IGF1R-CHK2-p53 signalling pathway.


Sujet(s)
Checkpoint kinase 2 , Diabète expérimental , Diabète de type 2 , Néphropathies diabétiques , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Rat Sprague-Dawley , Récepteur IGF de type 1 , Transduction du signal , Protéine p53 suppresseur de tumeur , Cordon ombilical , Animaux , Mâle , Rats , Récepteur IGF de type 1/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Humains , Diabète de type 2/métabolisme , Diabète de type 2/thérapie , Cordon ombilical/cytologie , Checkpoint kinase 2/métabolisme , Cellules souches mésenchymateuses/métabolisme , Néphropathies diabétiques/thérapie , Néphropathies diabétiques/métabolisme , Diabète expérimental/thérapie , Diabète expérimental/métabolisme , Alimentation riche en graisse , Altération de l'ADN , Glycémie/métabolisme
16.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891860

RÉSUMÉ

Osteoarthritis (OA) is a degenerative joint disease commonly found in elderly people and obese patients. Currently, OA treatments are determined based on their condition severity and a medical professional's advice. The aim of this study was to differentiate human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) into chondrocytes for transplantation in OA-suffering guinea pigs. hWJ-MSCs were isolated using the explant culture method, and then, their proliferation, phenotypes, and differentiation ability were evaluated. Subsequently, hWJ-MSCs-derived chondrocytes were induced and characterized based on immunofluorescent staining, qPCR, and immunoblotting techniques. Then, early-OA-suffering guinea pigs were injected with hyaluronic acid (HA) containing either MSCs or 14-day-old hWJ-MSCs-derived chondrocytes. Results showed that hWJ-MSCs-derived chondrocytes expressed specific markers of chondrocytes including Aggrecan, type II collagen, and type X collagen proteins and ß-catenin, Sox9, Runx2, Col2a1, Col10a1, and ACAN gene expression markers. Administration of HA plus hWJ-MSCs-derived chondrocytes (HA-CHON) produced a better recovery rate of degenerative cartilages than HA plus MSCs or only HA. Histological assessments demonstrated no significant difference in Mankin's scores of recovered cartilages between HA-CHON-treated guinea pigs and normal articular cartilage guinea pigs. Transplantation of hWJ-MSCs-derived chondrocytes was more effective than undifferentiated hWJ-MSCs or hyaluronic acid for OA treatment in guinea pigs. This study provides a promising treatment to be used in early OA patients to promote recovery and prevent disease progression to severe osteoarthritis.


Sujet(s)
Différenciation cellulaire , Chondrocytes , Modèles animaux de maladie humaine , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Arthrose , Cordon ombilical , Gelée de Wharton , Animaux , Cochons d'Inde , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Chondrocytes/métabolisme , Chondrocytes/cytologie , Arthrose/thérapie , Arthrose/anatomopathologie , Arthrose/métabolisme , Humains , Gelée de Wharton/cytologie , Transplantation de cellules souches mésenchymateuses/méthodes , Cordon ombilical/cytologie , Acide hyaluronique/pharmacologie , Cellules cultivées
17.
Placenta ; 153: 59-74, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38823320

RÉSUMÉ

INTRODUCTION: Preeclampsia (PE) is a pregnancy-specific complication. Its etiology and pathogenesis remain unclear. Previous studies have shown that neutrophil extracellular traps (NETs) cause placental dysfunction and lead to PE. Human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-EXOs) have been widely used to treat different diseases. We investigated whether hUCMSC-EXOs can protect against NET-induced placental damage. METHODS: NETs were detected in the placenta by immunofluorescence. The impact of NETs on cellular function and the effect of hUCMSC-EXOs on NET-induced placental damage were evaluated by 5-ethynyl-20-deoxyuridine (EdU) cell proliferation, lactate dehydrogenase (LDH), reactive oxygen species (ROS), and cell migration, invasion and tube formation assays; flow cytometry; and Western blotting. RESULTS: The number of placental NETs was increased in PE patients compared with control individuals. NETs impaired the function of endothelial cells and trophoblasts. These effects were partially reversed after N-acetyl-L-cysteine (NAC; ROS inhibitor) or DNase I (NET lysing agent) pretreatment. HUCMSC-EXOs ameliorated NET-induced functional impairment of endothelial cells and trophoblasts in vitro, partially reversed NET-induced inhibition of endothelial cell and trophoblast proliferation, and partially restored trophoblast migration and invasion and endothelial cell tube formation. Exosomes inhibited ROS production in these two cell types, suppressed p38 mitogen-activated protein kinase (p38 MAPK) signaling activation, activated extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, and modulated the Bax, Bim, Bcl-2 and cleaved caspase-3 levels to inhibit apoptosis. DISCUSSION: HUCMSC-EXOs can reverse NET-induced placental endothelial cell and trophoblast damage, possibly constituting a theoretical basis for the treatment of PE with exosomes.


Sujet(s)
Exosomes , Pièges extracellulaires , Cellules souches mésenchymateuses , Placenta , Pré-éclampsie , Cordon ombilical , Humains , Exosomes/métabolisme , Femelle , Grossesse , Pièges extracellulaires/métabolisme , Cellules souches mésenchymateuses/métabolisme , Placenta/métabolisme , Cordon ombilical/cytologie , Cordon ombilical/métabolisme , Pré-éclampsie/métabolisme , Adulte , Trophoblastes/métabolisme , Espèces réactives de l'oxygène/métabolisme
18.
Clin Exp Hypertens ; 46(1): 2366270, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38864268

RÉSUMÉ

OBJECTIVE: To elucidate the underlying mechanism by which the proliferation and migration abilities of human umbilical cord mesenchymal stem cells (hUC-MSCs) determine their therapeutic efficacy in rheumatoid arthritis treatment. METHODS: The DBA/1J mice were utilized to establish a collagen-induced RA (CIA) mouse model and to validate the therapeutic efficacy of hUC-MSCs transfected with CD151 siRNA. RNA-seq, QT-PCR and western blotting were utilized to evaluate the mRNA and protein levels of the PI3K/AKT pathway, respectively. RESULTS: IFN-γ significantly enhanced the proliferation and migration abilities of hUC-MSCs, up-regulating the expression of CD151, a gene related to cell proliferation and migration. Effective inhibition of this effect was achieved through CD151 siRNA treatment. However, IFN-γ did not affect hUC-MSCs differentiation or changes in cell surface markers. Additionally, transplantation of CD151-interfered hUC-MSCs (siRNA-CD151-hUC-MSCs) resulted in decreased colonization in the toes of CIA mice and worse therapeutic effects compared to empty vector treatment (siRNA-NC-hUC-MSCs). CONCLUSION: IFN-γ facilitates the proliferation and migration of hUC-MSCs through the CD151/PI3K/AKT pathway. The therapeutic efficacy of siRNA-CD151-hUC-MSCs was found to be inferior to that of siRNA-NC-hUC-MSCs.


Sujet(s)
Polyarthrite rhumatoïde , Mouvement cellulaire , Prolifération cellulaire , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Souris de lignée DBA , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/métabolisme , Souris , Cellules souches mésenchymateuses/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Phosphatidylinositol 3-kinases/métabolisme , Humains , Interféron gamma/métabolisme , Cordon ombilical/cytologie , Arthrite expérimentale/thérapie , Arthrite expérimentale/métabolisme , Mâle
19.
Int J Nanomedicine ; 19: 4923-4939, 2024.
Article de Anglais | MEDLINE | ID: mdl-38828201

RÉSUMÉ

Purpose: In recent years, exosomes have been proved to be used to treat many diseases. However, due to the lack of uniform quality control standards for exosomes, the safety of exosomes is still a problem to be solved, especially now more and more exosomes are used in clinical trials, and its non-clinical safety evaluation is particularly important. However, there is no safety evaluation standard for exosomes at present. Therefore, this study will refer to the evaluation criteria of therapeutic biological products, adopt non-human primates to evaluate the non-clinical safety of human umbilical cord mesenchymal stem cell exosomes from the general pharmacology and immunotoxicity, aiming at establishing a safety evaluation system of exosomes and providing reference for the clinical application of exosomes in the future. Methods: 3.85 × 1012 exosomes derived from human umbilical cord mesenchymal stem cells were injected into cynomolgus monkeys intravenously. The changes of general clinical conditions, hematology, immunoglobulin, Th1/Th2 cytokines, T lymphocytes and B lymphocytes, and immune organs were observed before and within 14 days after injection. Results: The results showed that exosomes did not have obvious pathological effects on the general clinical conditions, blood, coagulation function, organ coefficient, immunoglobulin, Th1/Th2 cytokines, lymphocytes, major organs, and major immune organs (spleen, thymus, bone marrow) of cynomolgus monkeys. However, the number of granulocyte-macrophage colonies in exosomes group was significantly higher than that in control group. Conclusion: To sum up, the general pharmacological results and immunotoxicity results showed that the injection of 3.85 × 1012 exosomes may have no obvious adverse reactions to cynomolgus monkeys. This dose of exosomes is relatively safe for treatment, which provides basis research for non-clinical safety evaluation of exosomes and provides reliable research basis for future clinical application of exosomes.


Sujet(s)
Exosomes , Macaca fascicularis , Cellules souches mésenchymateuses , Cordon ombilical , Animaux , Exosomes/composition chimique , Cellules souches mésenchymateuses/cytologie , Humains , Cordon ombilical/cytologie , Mâle , Femelle , Cytokines/métabolisme
20.
Mol Biol Rep ; 51(1): 762, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874690

RÉSUMÉ

BACKGROUND: Hepatocellular carcinoma (HCC) is experiencing a concerning rise in both incidence and mortality rates. Current therapeutic strategies are limited in their effectiveness, largely due to the complex causes of the disease and significant levels of drug resistance. Given the latest developments in human umbilical cord mesenchymal stem cells (hUC-MSCs) research, there is a debate over the continued use of stem cell transplantation for treating tumors. Consequently, this study seeks to explore the role of hUC-MSCs in the management of HCC. METHODS AND RESULTS: HUC-MSCs increased the number (10.75 ± 1.50) in the DEN/TCPOBOP-induced mice hepatoma model, compared with DMSO group (7.25 ± 1.71). Moreover, the liver index in hUC-MSCs group (0.21 ± 0.06) was greater than that in DMSO group (0.09 ± 0.01). Immunohistochemical (IHC) analysis revealed that while hUC-MSCs did not alter Foxp3 expression, they significantly stimulated Ki67 expression, indicative of increased tumor cellular proliferation. Additionally, immunofluorescence (IF) studies showed that hUC-MSCs increased CD8+ T cell counts without affecting macrophage numbers. Notably, granzyme B expression remained nearly undetectable. We observed that serum IL-18 levels were higher in the hUC-MSCs group (109.66 ± 0.38 pg/ml) compared to the DMSO group (91.14 ± 4.37 pg/ml). Conversely, IL-1ß levels decreased in the hUC-MSCs group (63.00 ± 0.53 pg/ml) relative to the DMSO group (97.38 ± 9.08 pg/ml). CONCLUSIONS: According to this study, hUC-MSCs promoted the growth of liver tumors. Therefore, we proposed that hUC-MSCs are not suitable for treating HCC, as they exhibit clinically prohibited abnormalities.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Interleukine-18 , Tumeurs du foie , Cellules souches mésenchymateuses , Cordon ombilical , Cellules souches mésenchymateuses/métabolisme , Humains , Animaux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/métabolisme , Cordon ombilical/cytologie , Interleukine-18/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/thérapie , Souris , Transplantation de cellules souches mésenchymateuses/méthodes , Mâle , Lignée cellulaire tumorale , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE