Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 15.915
Filtrer
1.
Ren Fail ; 46(2): 2381597, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39039856

RÉSUMÉ

BACKGROUND AND AIMS: Diabetic kidney disease (DKD) is one of the most common complications of diabetes. It is reported that mesenchymal stem cells (MSCs) derived exosomes (MSCs-Exo) may have great clinical application potential for the treatment of DKD, but the underlying mechanism has not been illustrated. To clarify the effect of MSC-Exo on NOD2 signaling pathway in podocytes under high glucose (HG) and DKD, we conduct this study. METHODS: We co-cultured podocytes and MSCs-Exo under 30 mM HG and injected MSCs-Exo into DKD mice, then we detected the NOD2 signaling pathway by western blot, qRT-PCT, immunofluorescence, transmission electron microscopy and immunohistochemistry both in vitro and in vivo. RESULTS: In vitro, HG lead to the apoptosis, increased the ROS level and activated the NOD2 signaling pathway in podocytes, while MSCs-Exo protected podocytes from injury reduced the expression of inflammatory factors including TNF-α, IL-6, IL-1ß, and IL-18 and alleviated the inflammatory response, inhibited the activation of NOD2 signaling pathway and the expression of it's downstream protein p-P65, p-RIP2, prevented apoptosis, increased cell viability in podocytes caused by HG. In vivo, MSCs-Exo alleviated renal injury in DKD mice, protected renal function, decreased urinary albumin excretion and inhibited the activation of NOD2 signaling pathway as well as the inflammation in renal tissue. CONCLUSION: MSCs-Exo protected the podocytes and DKD mice from inflammation by mediating NOD2 pathway, MSCs-Exo may provide a new target for the treatment of DKD.


Sujet(s)
Apoptose , Néphropathies diabétiques , Exosomes , Cellules souches mésenchymateuses , Protéine adaptatrice de signalisation NOD2 , Podocytes , Transduction du signal , Animaux , Exosomes/métabolisme , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/thérapie , Cellules souches mésenchymateuses/métabolisme , Souris , Protéine adaptatrice de signalisation NOD2/métabolisme , Podocytes/métabolisme , Podocytes/anatomopathologie , Mâle , Souris de lignée C57BL , Glucose/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinase 2/métabolisme , Techniques de coculture , Transplantation de cellules souches mésenchymateuses/méthodes , Espèces réactives de l'oxygène/métabolisme , Rein/anatomopathologie , Rein/métabolisme , Diabète expérimental/complications
2.
Ren Fail ; 46(2): 2319330, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39049729

RÉSUMÉ

AIM: This study explores the possible therapeutic role of rats and mice bone marrow-derived mesenchymal stem cells (BM-MSCs) on renal damage and toxicity brought on by carbon tetrachloride (CCl4) in Wistar rats. METHODS: Following an intraperitoneal injection of CCl4 (0.5 mL/kg b.w. twice weekly) for eight weeks, male Wistar rats were intravenously treated with rats and mice BM-MSCs (1 × 106 cells in 0.2 mL Dulbecco's Modified Eagle Medium (DMEM)/rat/week) a week for four weeks. Kidney functions were evaluated and kidney samples were examined using hematoxylin and eosin (H&E), Masson's trichrome (MT) staining techniques, and electron microscopy analysis. Kidney cyclooxygenase-2 (COX-2), protein 53 (p53), and tumor necrosis factor-α (TNF-α) were detected by immunohistochemical staining techniques. Additionally, bioindicators of oxidative stress and antioxidant defense systems were identified in kidney tissue. RESULTS: In CCl4-injected rats, serum creatinine, urea, and uric acid levels significantly increased, as did renal lipid peroxidation (LPO), while superoxide dismutase, glutathione peroxidase (GPx), glutathione (GSH) transferase, and GSH levels significantly dropped in the kidneys. Histologically, the kidneys displayed a wide range of structural abnormalities, such as glomerular shrinkage, tubular dilations, inflammatory leukocytic infiltration, fibroblast proliferation, and elevated collagen content. Inflammatory cytokines like COX-2 and TNF-α as well as the pro-apoptotic mediator p53 were considerably upregulated. Treatment of BM-MSCs from mice and rats with CCl4-injected rats considerably reduced the previously noted abnormalities. CONCLUSIONS: By boosting antioxidant defense and reducing apoptosis and inflammation, BM-MSCs from mice and rats were able to enhance kidney function and histological integrity in rats that had received CCl4 injections.


Sujet(s)
Tétrachloro-méthane , Fibrose , Rein , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Stress oxydatif , Rat Wistar , Animaux , Mâle , Tétrachloro-méthane/toxicité , Rats , Rein/anatomopathologie , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/métabolisme , Souris , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/thérapie , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/induit chimiquement , Cyclooxygenase 2/métabolisme , Peroxydation lipidique , Facteur de nécrose tumorale alpha/métabolisme , Modèles animaux de maladie humaine
4.
Mol Cell Biochem ; 479(7): 1643-1671, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38977625

RÉSUMÉ

Multiple sclerosis (MS) is a chronic neurodegenerative, inflammatory, and demyelinating disease of the central nervous system (CNS). Current medicines are not sufficient to control the inflammation and progressive damage to the CNS that is known in MS. These drawbacks highlight the need for novel treatment options. Cell therapy can now be used to treat complex diseases when conventional therapies are ineffective. Mesenchymal stem cells (MSCs) are a diverse group of multipotential non-hematopoietic stromal cells which have immunomodulatory, neurogenesis, and remyelinating capacity. Their advantageous effects mainly rely on paracrine, cell-cell communication and differentiation properties which introduced them as excellent candidates for MS therapy. Exosomes, as one of the MSCs secretomes, have unique properties that make them highly promising candidates for innovative approach in regenerative medicine. This review discusses the therapeutic potential of MSCs and their derived exosomes as a novel treatment for MS, highlighting the differences between these two approaches.


Sujet(s)
Exosomes , Cellules souches mésenchymateuses , Sclérose en plaques , Humains , Cellules souches mésenchymateuses/métabolisme , Exosomes/métabolisme , Sclérose en plaques/thérapie , Sclérose en plaques/anatomopathologie , Sclérose en plaques/métabolisme , Transplantation de cellules souches mésenchymateuses , Animaux , Communication cellulaire
5.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000343

RÉSUMÉ

Mesenchymal stem cells (MSCs) have shown great potential for the treatment of liver injuries, and the therapeutic efficacy greatly depends on their homing to the site of injury. In the present study, we detected significant upregulation of hepatocyte growth factor (HGF) in the serum and liver in mice with acute or chronic liver injury. In vitro study revealed that upregulation of miR-9-5p or miR-221-3p promoted the migration of human MSCs (hMSCs) toward HGF. Moreover, overexpression of miR-9-5p or miR-221-3p promoted hMSC homing to the injured liver and resulted in significantly higher engraftment upon peripheral infusion. hMSCs reduced hepatic necrosis and inflammatory infiltration but showed little effect on extracellular matrix (ECM) deposition. By contrast, hMSCs overexpressing miR-9-5p or miR-221-3p resulted in not only less centrilobular necrosis and venous congestion but also a significant reduction of ECM deposition, leading to obvious improvement of hepatocyte morphology and alleviation of fibrosis around central vein and portal triads. Further studies showed that hMSCs inhibited the activation of hepatic stellate cells (HSCs) but could not decrease the expression of TIMP-1 upon acute injury and the expression of MCP-1 and TIMP-1 upon chronic injury, while hMSCs overexpressing miR-9-5p or miR-221-3p led to further inactivation of HSCs and downregulation of all three fibrogenic and proinflammatory factors TGF-ß, MCP-1, and TIMP-1 upon both acute and chronic injuries. Overexpression of miR-9-5p or miR-221-3p significantly downregulated the expression of α-SMA and Col-1α1 in activated human hepatic stellate cell line LX-2, suggesting that miR-9-5p and miR-221-3p may partially contribute to the alleviation of liver injury by preventing HSC activation and collagen expression, shedding light on improving the therapeutic efficacy of hMSCs via microRNA modification.


Sujet(s)
Cellules étoilées du foie , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , microARN , microARN/génétique , microARN/métabolisme , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules étoilées du foie/métabolisme , Animaux , Souris , Transplantation de cellules souches mésenchymateuses/méthodes , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/thérapie , Lésions hépatiques dues aux substances/génétique , Mâle , Tétrachloro-méthane/effets indésirables , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/génétique , Souris de lignée C57BL , Mouvement cellulaire
6.
J Transl Med ; 22(1): 663, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010157

RÉSUMÉ

The T-helper 17 (Th17) cell and regulatory T cell (Treg) axis plays a crucial role in the development of multiple sclerosis (MS), which is regarded as an immune imbalance between pro-inflammatory cytokines and the maintenance of immune tolerance. Mesenchymal stem cell (MSC)-mediated therapies have received increasing attention in MS research. In MS and its animal model experimental autoimmune encephalomyelitis, MSC injection was shown to alter the differentiation of CD4+T cells. This alteration occurred by inducing anergy and reduction in the number of Th17 cells, stimulating the polarization of antigen-specific Treg to reverse the imbalance of the Th17/Treg axis, reducing the inflammatory cascade response and demyelination, and restoring an overall state of immune tolerance. In this review, we summarize the mechanisms by which MSCs regulate the balance between Th17 cells and Tregs, including extracellular vesicles, mitochondrial transfer, metabolic reprogramming, and autophagy. We aimed to identify new targets for MS treatment using cellular therapy by analyzing MSC-mediated Th17-to-Treg polarization.


Sujet(s)
Tolérance immunitaire , Cellules souches mésenchymateuses , Sclérose en plaques , Lymphocytes T régulateurs , Cellules Th17 , Humains , Cellules Th17/immunologie , Lymphocytes T régulateurs/immunologie , Cellules souches mésenchymateuses/immunologie , Animaux , Sclérose en plaques/immunologie , Sclérose en plaques/thérapie , Transplantation de cellules souches mésenchymateuses
7.
Stem Cell Res Ther ; 15(1): 208, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992782

RÉSUMÉ

BACKGROUND: Mesenchymal stromal cells (MSCs) tropism for tumours allows their use as carriers of antitumoural factors and in vitro transcribed mRNA (IVT mRNA) is a promising tool for effective transient expression without insertional mutagenesis risk. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine with antitumor properties by stimulating the specific immune response. The aim of this work was to generate modified MSCs by IVT mRNA transfection to overexpress GM-CSF and determine their therapeutic effect alone or in combination with doxorubicin (Dox) in a murine model of hepatocellular carcinoma (HCC). METHODS: DsRed or GM-CSF IVT mRNAs were generated from a cDNA template designed with specific primers followed by reverse transcription. Lipofectamine was used to transfect MSCs with DsRed (MSC/DsRed) or GM-CSF IVT mRNA (MSC/GM-CSF). Gene expression and cell surface markers were determined by flow cytometry. GM-CSF secretion was determined by ELISA. For in vitro experiments, the J774 macrophage line and bone marrow monocytes from mice were used to test GM-CSF function. An HCC model was developed by subcutaneous inoculation (s.c.) of Hepa129 cells into C3H/HeN mice. After s.c. injection of MSC/GM-CSF, Dox, or their combination, tumour size and mouse survival were evaluated. Tumour samples were collected for mRNA analysis and flow cytometry. RESULTS: DsRed expression by MSCs was observed from 2 h to 15 days after IVT mRNA transfection. Tumour growth remained unaltered after the administration of DsRed-expressing MSCs in a murine model of HCC and MSCs expressing GM-CSF maintained their phenotypic characteristic and migration capability. GM-CSF secreted by modified MSCs induced the differentiation of murine monocytes to dendritic cells and promoted a proinflammatory phenotype in the J774 macrophage cell line. In vivo, MSC/GM-CSF in combination with Dox strongly reduced HCC tumour growth in C3H/HeN mice and extended mouse survival in comparison with individual treatments. In addition, the tumours in the MSC/GM-CSF + Dox treated group exhibited elevated expression of proinflammatory genes and increased infiltration of CD8 + T cells and macrophages. CONCLUSIONS: Our results showed that IVT mRNA transfection is a suitable strategy for obtaining modified MSCs for therapeutic purposes. MSC/GM-CSF in combination with low doses of Dox led to a synergistic effect by increasing the proinflammatory tumour microenvironment, enhancing the antitumoural response in HCC.


Sujet(s)
Carcinome hépatocellulaire , Doxorubicine , Facteur de stimulation des colonies de granulocytes et de macrophages , Tumeurs du foie , Cellules souches mésenchymateuses , ARN messager , Animaux , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Cellules souches mésenchymateuses/métabolisme , Souris , Tumeurs du foie/thérapie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , ARN messager/métabolisme , ARN messager/génétique , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Facteur de stimulation des colonies de granulocytes et de macrophages/génétique , Facteur de stimulation des colonies de granulocytes et de macrophages/métabolisme , Lignée cellulaire tumorale , Transplantation de cellules souches mésenchymateuses/méthodes , Humains , Souris de lignée C3H , Transfection
8.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 732-737, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38949143

RÉSUMÉ

Temporomandibular joint osteoarthritis (TMJOA) is a kind of organic disease with synovial inflammation, cartilage degeneration and subchondral bone remodeling as the main pathological changes. The current treatment is mainly to relieve symptoms, but cannot completely stop the progression of the disease. Mesenchymal stem cells (MSC) have multi-lineage differentiation potential and have good prospects in the repair therapy of TMJOA. Intra-articular injection of MSC from bone marrow, adipose, umbilical cord, dental pulp, etc. has been shown to be effective in numerous animal studies. The above exogenous MSCs can also be used as seed cells to participate in tissue engineering and repair more severe defects. Recent studies have shown that exosomes are important mediators of MSC action and have some potential in the treatment of TMJOA. As the mechanisms of TMJOA are further investigated, there is some prospect that endogenous repair capacity can be activated by local injection of relevant drugs targeting the resident stem cells in the joint.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Arthrose , Articulation temporomandibulaire , Ingénierie tissulaire , Arthrose/thérapie , Cellules souches mésenchymateuses/cytologie , Humains , Articulation temporomandibulaire/anatomopathologie , Animaux , Ingénierie tissulaire/méthodes , Injections articulaires , Différenciation cellulaire , Exosomes , Troubles de l'articulation temporomandibulaire/thérapie
9.
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
10.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000084

RÉSUMÉ

Despite the enormous efforts of the pharmaceutical industry in the generation of new drugs (55 new ones last year) [...].


Sujet(s)
Vieillissement , Cellules souches mésenchymateuses , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Transplantation de cellules souches mésenchymateuses/méthodes , Animaux
11.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000105

RÉSUMÉ

This study aims to evaluate and compare cellular therapy with human Wharton's jelly (WJ) mesenchymal stem cells (MSCs) and neural precursors (NPs) in experimental autoimmune encephalomyelitis (EAE), a preclinical model of Multiple Sclerosis. MSCs were isolated from WJ by an explant technique, differentiated to NPs, and characterized by cytometry and immunocytochemistry analysis after ethical approval. Forty-eight rats were EAE-induced by myelin basic protein and Freund's complete adjuvant. Forty-eight hours later, the animals received intraperitoneal injections of 250 ng/dose of Bordetella pertussis toxin. Fourteen days later, the animals were divided into the following groups: a. non-induced, induced: b. Sham, c. WJ-MSCs, d. NPs, and e. WJ-MSCs plus NPs. 1 × 105. Moreover, the cells were placed in a 10 µL solution and injected via a stereotaxic intracerebral ventricular injection. After ten days, the histopathological analysis for H&E, Luxol, interleukins, and CD4/CD8 was carried out. Statistical analyses demonstrated a higher frequency of clinical manifestation in the Sham group (15.66%) than in the other groups; less demyelination was seen in the treated groups than the Sham group (WJ-MSCs, p = 0.016; NPs, p = 0.010; WJ-MSCs + NPs, p = 0.000), and a lower cellular death rate was seen in the treated groups compared with the Sham group. A CD4/CD8 ratio of <1 showed no association with microglial activation (p = 0.366), astrocytes (p = 0.247), and cell death (p = 0.577) in WJ-MSCs. WJ-MSCs and NPs were immunomodulatory and neuroprotective in cellular therapy, which would be translated as an adjunct in demyelinating diseases.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Sclérose en plaques , Animaux , Encéphalomyélite auto-immune expérimentale/thérapie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Rats , Sclérose en plaques/thérapie , Sclérose en plaques/anatomopathologie , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Humains , Femelle , Thérapie cellulaire et tissulaire/méthodes , Cellules souches neurales , Modèles animaux de maladie humaine , Gelée de Wharton/cytologie
12.
Biochem Biophys Res Commun ; 727: 150313, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38954981

RÉSUMÉ

Sepsis, a life-threatening condition resulting in multiple organ dysfunction, is characterized by a dysregulated immune response to infection. Current treatment options are limited, leading to unsatisfactory outcomes for septic patients. Here, we present a series of studies utilizing compact bone mesenchymal stem cells (CB-MSCs) and their derived paracrine mediators, especially exosome (CB-MSCs-Exo), to treat mice with cecal ligation and puncture-induced sepsis. Our results demonstrate that CB-MSCs treatment significantly improves the survival rate of septic mice by mitigating excessive inflammatory response and attenuating sepsis-induced organ injuries. Furthermore, CB-MSCs-conditioned medium, CB-MSCs secretome (CB-MSCs-Sec), and CB-MSCs-Exo exhibit potent anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated murine macrophage (RAW264.7). Intriguingly, intravenous administration of CB-MSCs-Exo confers superior protection against inflammation and organ damage in septic mice compared to CB-MSCs in certain aspects. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS) shotgun proteomic analysis, we identify a range of characterized proteins derived from the paracrine activity of CB-MSCs, involved in critical biological processes such as immunomodulation and apoptosis. Our findings highlight that the paracrine products of CB-MSCs could serve as a promising cell-free therapeutic agent for sepsis.


Sujet(s)
Exosomes , Cellules souches mésenchymateuses , Souris de lignée C57BL , Communication paracrine , Sepsie , Animaux , Sepsie/thérapie , Sepsie/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Souris , Cellules RAW 264.7 , Exosomes/métabolisme , Exosomes/transplantation , Mâle , Transplantation de cellules souches mésenchymateuses/méthodes , Lipopolysaccharides , Milieux de culture conditionnés/pharmacologie
13.
Front Immunol ; 15: 1400533, 2024.
Article de Anglais | MEDLINE | ID: mdl-39015561

RÉSUMÉ

Interleukin-6 (IL-6) is a versatile cytokine crucial for immune response modulation, inflammation regulation, and various physiological processes in the body. Its wide-ranging functions underscore its importance in maintaining health. Dysregulated IL-6 is closely associated with many diseases, making it a key research and therapeutic target. Elevated IL-6 levels in the central nervous system worsen neuroinflammation in neurodegenerative diseases by activating microglia and astrocytes and releasing pro-inflammatory cytokines and neurotoxic molecules. Moreover, dysregulated IL-6 weakens the blood-brain barrier, exacerbating neuroinflammation and neuronal damage by allowing peripheral immune cells and inflammatory mediators to enter the brain. Mesenchymal stem cells (MSCs) show promise in modulating neuroinflammation by regulating IL-6 levels. They effectively suppress pro-inflammatory cytokines, including IL-6, while promoting anti-inflammatory factors. This therapeutic approach highlights the importance of targeting IL-6 and other inflammatory mediators to alleviate neuroinflammation and its adverse effects on neurological disorders. This review provides a comprehensive overview of IL-6's involvement in neurological disorders, examining endogenous IL-6 and IL-6 derived from MSCs. We explore IL-6's mechanisms affecting neuronal function, survival, and immune modulation in the central nervous system. Additionally, we discuss the potential of MSC-derived IL-6 in neuroregeneration and neuroprotection. By elucidating IL-6's interplay with neurological pathologies, this review offers insights into novel therapeutic strategies targeting IL-6 signaling pathways for neurological disorders.


Sujet(s)
Interleukine-6 , Cellules souches mésenchymateuses , Animaux , Humains , Interleukine-6/métabolisme , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/immunologie , Cellules souches mésenchymateuses/métabolisme , Maladies du système nerveux/thérapie , Maladies du système nerveux/immunologie , Maladies du système nerveux/métabolisme , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/thérapie , Transduction du signal
15.
Stem Cell Res Ther ; 15(1): 199, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38971781

RÉSUMÉ

BACKGROUND: Bone marrow-derived mesenchymal stem cell (BMMSC)-based therapy has become a major focus for treating liver fibrosis/cirrhosis. However, although these cell therapies promote the treatment of this disease, the heterogeneity of BMMSCs, which causes insufficient efficacy during clinical trials, has not been addressed. In this study, we describe a novel Percoll-Plate-Wait procedure (PPWP) for the isolation of an active cell subset from BMMSC cultures that was characterized by the expression of neuroglial antigen 2 (NG2/BMMSCs). METHODS: By using the key method of PPWP and other classical biological techniques we compared NG2/BMMSCs with parental BMMSCs in biological and functional characteristics within a well-defined diethylnitrosamine (DEN)-induced liver fibrosis/cirrhosis injury male C57BL/6 mouse model also in a culture system. Of note, the pathological alterations in the model is quite similar to humans'. RESULTS: The NG2/BMMSCs revealed more advantages compared to parentalBMMSCs. They exhibited greater proliferation potential than parental BMMSCs, as indicated by Ki-67 immunofluorescence (IF) staining. Moreover, higher expression of SSEA-3 (a marker specific for embryonic stem cells) was detected in NG2/BMMSCs than in parental BMMSCs, which suggested that the "stemness" of NG2/BMMSCs was greater than that of parental BMMSCs. In vivo studies revealed that an injection of NG2/BMMSCs into mice with ongoing DEN-induced liver fibrotic/cirrhotic injury enhanced repair and functional recovery to a greater extent than in mice treated with parental BMMSCs. These effects were associated with the ability of NG2/BMMSCs to differentiate into bile duct cells (BDCs). In particular, we discovered for the first time that NG2/BMMSCs exhibit unique characteristics that differ from those of parental BMMSCs in terms of producing liver sinusoidal endothelial cells (LSECs) to reconstruct injured blood vessels and sinusoidal structures in the diseased livers, which are important for initiating hepatocyte regeneration. This unique potential may also suggest that NG2/BMMSCs could be an novel off-liver progenitor of LSECs. Ex vivo studies revealed that the NG2/BMMSCs exhibited a similar trend to that of their in vivo in terms of functional differentiation responding to the DEN-diseased injured liver cues. Additionally, the obvious core role of NG2/BMMSCs in supporting the functions of BMMSCs in bile duct repair and BDC-mediated hepatocyte regeneration might also be a novel finding. CONCLUSIONS: Overall, the PPWP-isolated NG2/BMMSCs could be a novel effective cell subset with increased purity to serve as a new therapeutic tool for enhancing treatment efficacy of BMMSCs and special seed cell source (BDCs, LSECs) also for bioliver engineering.


Sujet(s)
Antigènes , Cirrhose du foie , Cellules souches mésenchymateuses , Souris de lignée C57BL , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Cirrhose du foie/thérapie , Cirrhose du foie/anatomopathologie , Cirrhose du foie/induit chimiquement , Souris , Mâle , Antigènes/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Protéoglycanes/métabolisme , Différenciation cellulaire , Prolifération cellulaire , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/métabolisme , Cellules cultivées
16.
Stem Cell Res Ther ; 15(1): 215, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39020413

RÉSUMÉ

BACKGROUND: A favorable regenerative microenvironment is essential for peripheral nerve regeneration. Neural tissue-specific extracellular matrix (ECM) is a natural material that helps direct cell behavior and promote axon regeneration. Both bone marrow-derived mesenchymal stem cells (BMSCs) and adipose-derived mesenchymal stem cells (ADSCs) transplantation are effective in repairing peripheral nerve injury (PNI). However, there is no study that characterizes the in vivo microenvironmental characteristics of these two MSCs for the early repair of PNI when combined with neural tissue-derived ECM materials, i.e., acellular nerve allograft (ANA). METHODS: In order to investigate biological characteristics, molecular mechanisms of early stage, and effectiveness of ADSCs- or BMSCs-injected into ANA for repairing PNI in vivo, a rat 10 mm long sciatic nerve defect model was used. We isolated primary BMSCs and ADSCs from bone marrow and adipose tissue, respectively. First, to investigate the in vivo response characteristics and underlying molecular mechanisms of ANA combined with BMSCs or ADSCs, eighty-four rats were randomly divided into three groups: ANA group, ANA+BMSC group, and ANA+ADSC group. We performed flow cytometry, RT-PCR, and immunofluorescence staining up to 4 weeks postoperatively. To further elucidate the underlying molecular mechanisms, changes in long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs) were systematically investigated using whole transcriptome sequencing. We then constructed protein-protein interaction networks to find 10 top ranked hub genes among differentially expressed mRNAs. Second, in order to explore the effectiveness of BMSCs and ADSCs on neural tissue-derived ECM materials for repairing PNI, sixty-eight rats were randomized into four groups: ANA group, ANA+BMSC group, ANA+ADSC group, and AUTO group. In the ANA+BMSC and ANA+ADSC groups, ADSCs/BMSCs were equally injected along the long axis of the 10-mm ANA. Then, we performed histological and functional assessments up to 12 weeks postoperatively. RESULTS: The results of flow cytometry and RT-PCR showed that ANA combined with BMSCs exhibited more significant immunomodulatory effects, as evidenced by the up-regulation of interleukin (IL)-10, down-regulation of IL-1ß and tumor necrosis factor-alpha (TNF-α) expression, promotion of M1-type macrophage polarization to M2-type, and a significant increase in the number of regulatory T cells (Tregs). ANA combined with ADSCs exhibited more pronounced features of pro-myelination and angiogenesis, as evidenced by the up-regulation of myelin-associated protein gene (MBP and MPZ) and angiogenesis-related factors (TGF-ß, VEGF). Moreover, differentially expressed genes from whole transcriptome sequencing results further indicated that ANA loaded with BMSCs exhibited notable immunomodulatory effects and ANA loaded with ADSCs was more associated with angiogenesis, axonal growth, and myelin formation. Notably, ANA infused with BMSCs or ADSCs enhanced peripheral nerve regeneration and motor function recovery with no statistically significant differences. CONCLUSIONS: This study revealed that both ANA combined with BMSCs and ADSCs enhance peripheral nerve regeneration and motor function recovery, but their biological characteristics (mainly including immunomodulatory effects, pro-vascular regenerative effects, and pro-myelin regenerative effects) and underlying molecular mechanisms in the process of repairing PNI in vivo are different, providing new insights into MSC therapy for peripheral nerve injury and its clinical translation.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Régénération nerveuse , Lésions des nerfs périphériques , Rat Sprague-Dawley , Ingénierie tissulaire , Animaux , Rats , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Ingénierie tissulaire/méthodes , Lésions des nerfs périphériques/thérapie , Lésions des nerfs périphériques/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Nerf ischiatique/traumatismes , Nerf ischiatique/métabolisme , Mâle , Tissu adipeux/cytologie , Tissu adipeux/métabolisme
17.
Stem Cell Res Ther ; 15(1): 211, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39020426

RÉSUMÉ

Pain is a very common and complex medical problem that has a serious impact on individuals' physical and mental health as well as society. Non-steroidal anti-inflammatory drugs and opioids are currently the main drugs used for pain management, but they are not effective in controlling all types of pain, and their long-term use can cause adverse effects that significantly impair patients' quality of life. Mesenchymal stem cells (MSCs) have shown great potential in pain treatment. However, limitations such as the low proliferation rate of MSCs in vitro and low survival rate in vivo restrict their analgesic efficacy and clinical translation. In recent years, researchers have explored various innovative approaches to improve the therapeutic effectiveness of MSCs in pain treatment. This article reviews the latest research progress of MSCs in pain treatment, with a focus on methods to enhance the analgesic efficacy of MSCs, including engineering strategies to optimize the in vitro culture environment of MSCs and to improve the in vivo delivery efficiency of MSCs. We also discuss the unresolved issues to be explored in future MSCs and pain research and the challenges faced by the clinical translation of MSC therapy, aiming to promote the optimization and clinical translation of MSC-based analgesia therapy.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Gestion de la douleur , Humains , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Gestion de la douleur/méthodes , Animaux , Analgésie/méthodes , Douleur/traitement médicamenteux
18.
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
19.
Immun Inflamm Dis ; 12(7): e1337, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39023421

RÉSUMÉ

OBJECTIVE: To investigate the effect of nasal mucosa-derived ectodermal mesenchymal stem cells (NM-EMSCs) on the inflammatory state of rats with chronic rhinosinusitis (CRS) and the underlying therapeutic mechanism. METHODS: NM-EMSCs were isolated and extracted to construct a rat model of CRS. Fifteen Sprague‒Dawley (SD) rats were randomly divided into three groups: CK + NS group rats were injected locally with saline in the nasal mucosa; CRS + NS group rats were injected locally with saline in the nasal mucosa; and CRS + EMSCs group rats were injected locally with NM-EMSCs in the nasal mucosa. One rat from the CRS + EMSCs group was randomly euthanized at 2, 4, and 6 days after injection, and the nasal mucosa tissues were collected for HE staining, Masson's trichrome staining, and periodic acid-Schiff staining. RESULTS: NM-EMSCs specifically expressing CD73, CD105, and CD90 were successfully isolated from the nasal mucosa of rats and were able to differentiate into adipocytes, osteoblasts, and chondrocytes. After saline and NM-EMSC injection, compared with those in the blank control CK + NS group, the nasal mucosa in the CRS + NS and CRS + EMSC groups exhibited obvious thickening, a large amount of inflammatory cell infiltration, and increased collagen and mucin distribution. Four days post-NM-EMSC injection, the thickening of the nasal mucosa in the CRS group was gradually alleviated, the inflammatory cell infiltration gradually decreased, and the distribution of collagen and mucin and the collagen-positive area gradually decreased. Moreover, only a small number of inflammatory cells were visible, and the distribution of mucins was limited to 6 days post-NM-EMSC injection. CONCLUSION: NM-EMSCs effectively attenuated inflammation in the nasal mucosa of CRS model rats.


Sujet(s)
Différenciation cellulaire , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Muqueuse nasale , Rat Sprague-Dawley , Rhinite , Sinusite , Animaux , Muqueuse nasale/métabolisme , Muqueuse nasale/anatomopathologie , Muqueuse nasale/immunologie , Sinusite/thérapie , Sinusite/immunologie , Sinusite/anatomopathologie , Rats , Cellules souches mésenchymateuses/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Maladie chronique , Rhinite/thérapie , Rhinite/immunologie , Rhinite/anatomopathologie , Modèles animaux de maladie humaine , Cellules cultivées , Mâle ,
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE