Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Adv Sci (Weinh) ; 10(27): e2300470, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37505480

RÉSUMÉ

Myocardial infarction (MI) causes excessive damage to the myocardium, including the epicardium. However, whether pluripotent stem cell-derived epicardial cells (EPs) can be a therapeutic approach for infarcted hearts remains unclear. Here, the authors report that intramyocardial injection of human embryonic stem cell-derived EPs (hEPs) at the acute phase of MI ameliorates functional worsening and scar formation in mouse hearts, concomitantly with enhanced cardiomyocyte survival, angiogenesis, and lymphangiogenesis. Mechanistically, hEPs suppress MI-induced infiltration and cytokine-release of inflammatory cells and promote reparative macrophage polarization. These effects are blocked by a type I interferon (IFN-I) receptor agonist RO8191. Moreover, intelectin 1 (ITLN1), abundantly secreted by hEPs, interacts with IFN-ß and mimics the effects of hEP-conditioned medium in suppression of IFN-ß-stimulated responses in macrophages and promotion of reparative macrophage polarization, whereas ITLN1 downregulation in hEPs cancels beneficial effects of hEPs in anti-inflammation, IFN-I response inhibition, and cardiac repair. Further, similar beneficial effects of hEPs are observed in a clinically relevant porcine model of reperfused MI, with no increases in the risk of hepatic, renal, and cardiac toxicity. Collectively, this study reveals hEPs as an inflammatory modulator in promoting infarct healing via a paracrine mechanism and provides a new therapeutic approach for infarcted hearts.


Sujet(s)
Cellules souches embryonnaires humaines , Infarctus du myocarde , Suidae , Souris , Humains , Animaux , Myocarde , Myocytes cardiaques , Infarctus du myocarde/traitement médicamenteux , Macrophages
2.
Mol Reprod Dev ; 89(7): 281-289, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35678749

RÉSUMÉ

The expansion of cumulus cells associated with oocytes is an essential phenomenon in normal mammalian ovulation. Indeed, attenuated expression of cumulus expansion-related genes, including Has2, Ptgs2, Ptx3, and Tnfaip6, results in ovulation failure, leading to female subfertility or infertility. Moreover, emerging evidence suggests that proteins of the fibroblast growth factor (FGF) family, produced within ovarian follicles, regulate the development and function of cumulus cells; however, the effects of FGF signaling on cumulus expansion have not been investigated extensively. Herein, we investigate the effects of FGF signaling, particularly those of FGF8 secreted by oocytes, on epidermal growth factor-induced cumulus expansion in mice. The phosphorylation level of MAPK3/1, an intracellular mediator of FGF signaling, was significantly decreased in cumulus-oocyte complexes (COCs) following treatment with NVP-BGJ398, an FGF receptor inhibitor. Moreover, even though NVP-BGJ398 treatment did not affect cumulus cell expansion, it significantly upregulated the expression of Ptgs2 and Ptx3. In contrast, treatment with recombinant FGF8 did not affect the degree of cumulus expansion or the expression of expansion-related genes in COCs or oocytectomized cumulus cell complexes. Collectively, these results suggest that FGFs, other than FGF8, exert suppressive effects on the cumulus expansion process in mice.


Sujet(s)
Cellules du cumulus , Facteurs de croissance fibroblastique , Animaux , Cellules du cumulus/métabolisme , Cyclooxygenase 2/génétique , Cyclooxygenase 2/métabolisme , Femelle , Facteur de croissance fibroblastique de type 8/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Mammifères , Souris , Ovocytes/métabolisme , Follicule ovarique/physiologie
3.
Zygote ; 30(5): 730-734, 2022 Oct.
Article de Anglais | MEDLINE | ID: mdl-35416145

RÉSUMÉ

This study evaluated the effect of fibroblast growth factor-2 (FGF-2) on the morphology, primordial follicle activation and growth after in vitro culture of domestic cat ovarian tissue. Ovaries (n = 12) from prepubertal domestic cats were collected and fragmented. One fragment was fixed for histological analysis (fresh control). The remaining fragments were incubated in control medium alone or with 10, 50 or 100 ng/ml FGF-2 for 7 days. After in vitro culture, the following endpoints were analyzed: morphology, activation by counting primordial and developing follicles, and growth (follicle and oocyte diameters). Treatment with 100 ng/ml FGF-2 maintained (P > 0.05) the percentage of normal follicles similar to fresh control. Follicle survival was greater (P < 0.05) after culture in 100 ng/ml FGF-2 than in 50 ng/ml FGF-2. The percentage of primordial follicles decreased (P < 0.05) and the percentage of developing follicles increased (P < 0.05) in all treatments compared with fresh tissue. The proportion of developing follicles increased (P < 0.05) in tissues incubated with 100 ng/ml FGF-2 compared with control medium and other FGF-2 concentrations. Furthermore, culture in 10 or 100 ng/ml FGF-2 resulted in increased (P < 0.05) follicle and oocyte diameters compared with fresh tissues and MEM+. In conclusion, FGF-2 at 100 ng/ml maintains follicle survival and promotes the in vitro activation and growth of cat primordial follicles.


Sujet(s)
Facteur de croissance fibroblastique de type 2 , Follicule ovarique , Animaux , Chats , Femelle , Facteur de croissance fibroblastique de type 2/pharmacologie , Ovocytes/physiologie , Follicule ovarique/physiologie , Ovaire , Techniques de culture de tissus/méthodes
4.
Curr Mol Med ; 22(2): 120-131, 2022.
Article de Anglais | MEDLINE | ID: mdl-33550972

RÉSUMÉ

Cell-based regenerative therapies involving stem or progenitor cells are considered as possible therapeutic modalities to treat non-communicable and degenerative diseases. Recently, regenerative outcomes of cell-based therapies have been linked to paracrine factors and extracellular vesicles [EVs] released by the transplanted cells rather than the transplanted cells themselves. EVs contain a cargo that includes microRNAs [miRNAs], mRNAs, as well as proteins. Their role in mediating intercellular communication has been acknowledged in several studies. However, the regenerative potential of the miRNAs, mRNAs, and proteins that are present in EVs is a matter of ongoing scientific debate. In this review, we discuss EVs as an alternative to stem cell-based therapy to treat some of the non-communicable and degenerative diseases. Moreover, we also propose that pre-treatment of the cells could help to produce EVs enriched with particular miRNAs, mRNAs, and/or proteins that could support the successful regeneration of a targeted organ.


Sujet(s)
Exosomes , Vésicules extracellulaires , Cellules souches mésenchymateuses , microARN , Exosomes/métabolisme , Vésicules extracellulaires/métabolisme , Cellules souches mésenchymateuses/métabolisme , microARN/génétique , microARN/métabolisme , Cellules souches/métabolisme
5.
Mater Sci Eng C Mater Biol Appl ; 124: 112077, 2021 May.
Article de Anglais | MEDLINE | ID: mdl-33947569

RÉSUMÉ

The heart contains a wide range of cell types, which are not isolated but interact with one another via multifarious paracrine, autocrine and endocrine factors. In terms of cardiac angiogenesis, previous studies have proved that regulating the communication between cardiomyocytes and endothelial cells is efficacious to promote capillary formation. Firstly, this study investigated the effect and underlying mechanism of bioactive glass (BG) acted on vascular endothelial growth factor (VEGF) paracrine signaling in cardiomyocytes. We found that bioactive ions released from BG significantly promoted the VEGF production and secretion of cardiomyocytes. Subsequently, we proved that cardiomyocyte-derived VEGF played an important role in mediating the behavior of endothelial cells. Further research showed that the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/hypoxia-inducible factor 1α (HIF-1α) signaling pathway was upregulated by BG, which was involved in VEGF expression of cardiomyocytes. This study revealed that by means of modulating cellular crosstalk via paracrine signaling of host cells in heart is a new direction for the application of BGs in cardiac angiogenesis.


Sujet(s)
Myocytes cardiaques , Facteur de croissance endothéliale vasculaire de type A , Cellules endothéliales , Sous-unité alpha du facteur-1 induit par l'hypoxie , Communication paracrine , Phosphatidylinositol 3-kinases
6.
Nano Converg ; 7(1): 34, 2020 Oct 16.
Article de Anglais | MEDLINE | ID: mdl-33064240

RÉSUMÉ

Cell therapy based on human adipose derived stem cells (hADSCs) is a known potential therapeutic approach to induce angiogenesis in ischemic diseases. However, the therapeutic efficacy of direct hADSC injection is limited by a low cell viability and poor cell engraftment after administration. To improve the outcomes of this kind of approach, various types of nanoparticles have been utilized to improve the therapeutic efficacy of hADSC transplantation. Despite their advantages, the adverse effects of nanoparticles, such as genetic damage and potential oncogenesis based on non-degradable property of nanoparticles prohibit the application of nanoparticles toward the clinical applications. Herein, we designed a transition metal based inorganic nanocluster able of pH-selective degradation (ps-TNC), with the aim of enhancing an hADSC based treatment of mouse hindlimb ischemia. Our ps-TNC was designed to undergo degradation at low pH conditions, thus releasing metal ions only after endocytosis, in the endosome. To eliminate the limitations of both conventional hADSC injection and non-degradable property of nanoparticles, we have collected conditioned medium (CM) from the ps-TNC treated hADSCs and administrated it to the ischemic lesions. We found that intracellular increment of transition metal ion upregulated the hypoxia-inducible factor 1α, which can induce vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) expressions. Based on the molecular mechanism, the secretion of VEGF and bFGF by ps-TNC treated hADSCs showed a significant improvement compared to that of untreated cells. Injecting the CM collected from ps-TNC treated hADSCs into the mouse hindlimb ischemia model (ps-TNC-CM group) showed significantly improved angiogenesis in the lesions, with improved limb salvage and decreased muscle degeneration compared to the group injected with CM collected from normal hADSCs (CM group). This study suggests a novel strategy, combining a known angiogenesis molecular mechanism with both an improvement on conventional stem cell therapy and the circumvention of some limitations still present in modern approaches based on nanoparticles.

7.
Int J Mol Sci ; 21(17)2020 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-32872523

RÉSUMÉ

Extracellular matrix (ECM) components play an important role in maintaining skeletal muscle function, but excessive accumulation of ECM components interferes with skeletal muscle regeneration after injury, eventually inducing fibrosis. Increased oxidative stress level caused by dystrophin deficiency is a key factor in fibrosis in Duchenne muscular dystrophy (DMD) patients. Mesenchymal stem cells (MSCs) are considered a promising therapeutic agent for various diseases involving fibrosis. In particular, the paracrine factors secreted by MSCs play an important role in the therapeutic effects of MSCs. In this study, we investigated the effects of MSCs on skeletal muscle fibrosis. In 2-5-month-old mdx mice intravenously injected with 1 × 105 Wharton's jelly (WJ)-derived MSCs (WJ-MSCs), fibrosis intensity and accumulation of calcium/necrotic fibers were significantly decreased. To elucidate the mechanism of this effect, we verified the effect of WJ-MSCs in a hydrogen peroxide-induced fibrosis myotubes model. In addition, we demonstrated that matrix metalloproteinase-1 (MMP-1), a paracrine factor, is critical for this anti-fibrotic effect of WJ-MSCs. These findings demonstrate that WJ-MSCs exert anti-fibrotic effects against skeletal muscle fibrosis, primarily via MMP-1, indicating a novel target for the treatment of muscle diseases, such as DMD.


Sujet(s)
Matrix Metalloproteinase 13/métabolisme , Cellules souches mésenchymateuses/métabolisme , Muscles squelettiques/cytologie , Myopathie de Duchenne/thérapie , Administration par voie intraveineuse , Animaux , Lignée cellulaire , Dipeptides/pharmacologie , Matrice extracellulaire/métabolisme , Femelle , Peroxyde d'hydrogène/effets indésirables , Transplantation de cellules souches mésenchymateuses , Souris , Souris de lignée mdx , Cellules musculaires/effets des médicaments et des substances chimiques , Cellules musculaires/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Grossesse , Inhibiteur tissulaire de métalloprotéinase-1/pharmacologie , Résultat thérapeutique
8.
J Sex Med ; 15(3): 284-295, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29502978

RÉSUMÉ

BACKGROUND: Whether combined transplantation of mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) is more effective than transplantation of a single cell type in the restoration of erectile function is unknown. AIM: To investigate the effect of combined transplantation of MSCs and EPCs on restoration of erectile function in rats with cavernous nerve injury (CNI). METHODS: MSCs were isolated from human bone marrow and EPCs were isolated from human umbilical cord blood. MSCs and EPCs were identified by flow cytometry and in vitro differentiation or immunofluorescence staining. 25 8-week-old male Sprague-Dawley rats were allocated to 1 of 5 groups: sham operation group, bilateral CNI group receiving periprostatic implantation of MSCs plus EPCs, MSCs, EPCs, or phosphate buffered saline (control group). 2 weeks after CNI and treatment, erectile function of rats was measured by electrically stimulating the CN. The penis and major pelvic ganglia were harvested for histologic examinations. RNA and protein levels of neurotrophin factors (vascular endothelial growth factor, nerve growth factor, and brain-derived neurotrophic factor) in mono- or coculture MSCs and EPCs were assessed by real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. OUTCOMES: Intracavernous pressure and mean arterial pressure were measured to evaluate erectile function. Histologic examinations of the penis and major pelvic ganglia and RNA and protein levels of neurotrophin factors in MSCs and EPCs were performed. RESULTS: MSCs and EPCs expressed the specified cell markers and exhibited the typical appearance and characteristics. Treatments using MSCs and/or EPCs could increase endothelial and smooth muscle contents of the corpus cavernosum, decrease caspase-3 expression and increase penile neuronal nitric oxide synthase expression, and restore the neural component of the major pelvic ganglia in rats with CNI. Combined transplantation of MSCs and EPCs had a better effect on improving erectile function than single transplantation of MSCs or EPCs. Expression levels of vascular endothelial growth factor and nerve growth factor in coculture MSCs and EPCs were significantly higher than those of primary MSCs or EPCs. CLINICAL TRANSLATION: Combined transplantation of MSCs and EPCs was more effective in restoring erectile function in CNI-related erectile dysfunction models. STRENGTHS AND LIMITATIONS: The study, for the 1st time, proved that combined transplantation of MSCs and EPCs was more effective in restoring erectile function in rats with CNI. The rat model might not represent the human condition. CONCLUSION: Combined periprostatic transplantation of MSCs and EPCs could restore erectile function in rats with CNI more effectively. MSCs might restore CN fibers by secreting neurotrophin factors such as vascular endothelial growth factor and nerve growth factor, and EPCs could enhance the paracrine activity of MSCs. Fang J-f, Huang X-n, Han X-y, et al. Combined Transplantation of Mesenchymal Stem Cells and Endothelial Progenitor Cells Restores Cavernous Nerve Injury-Related Erectile Dysfunction. J Sex Med 2018;15:284-295.


Sujet(s)
Progéniteurs endothéliaux/transplantation , Dysfonctionnement érectile/thérapie , Transplantation de cellules souches mésenchymateuses/méthodes , Animaux , Facteur neurotrophique dérivé du cerveau/métabolisme , Différenciation cellulaire , Modèles animaux de maladie humaine , Dysfonctionnement érectile/physiopathologie , Humains , Mâle , Muscles lisses/métabolisme , Érection du pénis/physiologie , Rats , Rat Sprague-Dawley , Traumatismes du système nerveux/complications , Facteur de croissance endothéliale vasculaire de type A/métabolisme
9.
Life Sci ; 193: 257-269, 2018 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-29107793

RÉSUMÉ

Paracrine factors, including growth factors and cytokines, released from cardiac myocytes following ß-adrenergic receptor (ß-AR) stimulation regulate cardiac fibroblasts. Activated cardiac fibroblasts have the ability to increase collagen synthesis, cell proliferation and myofibroblast differentiation, leading to cardiac fibrosis. However, it is unknown which ß-AR subtypes and signaling pathways mediate the upregulation of paracrine factors in cardiac myocytes. In this study, we demonstrated that sustained stimulation of ß-ARs significantly induced synthesis and secretion of growth factors, including connective tissue growth factor (CTGF) and vascular endothelial growth factor (VEGF), via the cAMP-dependent and protein kinase A (PKA)-dependent pathways. In addition, isoproterenol (ISO)-mediated synthesis and secretion of CTGF and VEGF through the ß1-AR and ß2-AR subtypes. Paracrine factors released by cardiac myocytes following sustained ß-AR stimulation are necessary for the induction of cell proliferation and synthesis of collagen I, collagen III and α-smooth muscle actin (α-SMA) in cardiac fibroblasts, confirming that ß-AR overstimulation of cardiac myocytes induces cardiac fibrosis by releasing several paracrine factors. These effects can be antagonized by ß-blockers, including atenolol, metoprolol, and propranolol. Thus, the use of ß-blockers may have beneficial effects on the treatment of myocardial fibrosis in patients with heart failure.


Sujet(s)
Myocytes cardiaques/effets des médicaments et des substances chimiques , Récepteurs bêta-adrénergiques/métabolisme , Antagonistes bêta-adrénergiques/pharmacologie , Animaux , Animaux nouveau-nés , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cytokines/effets des médicaments et des substances chimiques , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Coeur/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intercellulaire/métabolisme , Isoprénaline/pharmacologie , Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Culture de cellules primaires , Rats , Rat Sprague-Dawley , Récepteurs bêta-adrénergiques/physiologie , Récepteurs bêta-2 adrénergiques/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de croissance endothéliale vasculaire de type A/métabolisme
10.
ACS Appl Mater Interfaces ; 9(44): 38344-38355, 2017 Nov 08.
Article de Anglais | MEDLINE | ID: mdl-29043772

RÉSUMÉ

Cell therapy has been suggested as a treatment modality for ischemic diseases, but the poor survival and engraftment of implanted cells limit its therapeutic efficacy. To overcome such limitation, we used electrical stimulation (ES) derived from a wearable solar cell for inducing angiogenesis in ischemic tissue. ES enhanced the secretion of angiogenic growth factors and the migration of mesenchymal stem cells (MSCs), myoblasts, endothelial progenitor cells, and endothelial cells in vitro. In a mouse ischemic hindlimb model, ES generated by a solar cell and applied to the ischemic region promoted migration of MSCs toward the ischemic site and upregulated expression of angiogenic paracrine factors (vascular endothelial, basic fibroblast, and hepatocyte growth factors; and stromal cell-derived factor-1α). Importantly, solar cell-generated ES promoted the formation of capillaries and arterioles at the ischemic region, attenuated muscle necrosis and fibrosis, and eventually prevented loss of the ischemic limb. Solar cell ES therapy showed higher angiogenic efficacy than conventional MSC therapy. This study shows the feasibility of using solar cell ES as a novel treatment for therapeutic angiogenesis.

11.
Brain Behav Immun ; 57: 47-52, 2016 Oct.
Article de Anglais | MEDLINE | ID: mdl-27132057

RÉSUMÉ

Recent evidence has indicated that overweight/obese children may experience cognitive and immune dysfunction, but the underlying mechanisms responsible for the association between overweight/obesity, immune dysfunction, and cognition have yet to be established. The present study aimed to identify a novel link between obesity-induced immune system dysregulation and cognition in preadolescent children. A total of 27 male children (age: 8-10years) were recruited and separated by body mass index (BMI) into healthy weight (HW: 5th-84.9th percentile, n=16) and overweight/obese (OW: ⩾85th percentile, n=11) groups. Adiposity was assessed using dual energy X-ray absorptiometry (DXA), and aspects of executive function were assessed using the Woodcock-Johnson III Tests of Cognitive Abilities. Monocyte populations (CD14(+)CD16(-), CD14(+)CD16(+)) with and without expression of chemokine receptor type 2 (CCR2), and circulating progenitor cells (CPCs: CD34(+)CD45(dim)), in peripheral blood were quantified by flow cytometry. CPCs were isolated by flow sorting and cultured for 24h for collection of conditioned media (CM) that was applied to SH-SY5Y neuroblastomas to examine the paracrine effects of CPCs on neurogenesis. OW had significantly higher quantities of both populations of monocytes (CD14(+)CD16(-): 57% increase; CD14(+)CD16(+): 95% increase, both p<0.01), monocytes expressing CCR2 (CD14(+)CD16(-)CCR2(+): 66% increase; CD14(+)CD16(+)CCR2(+): 168% increase, both p<0.01), and CPCs (47% increase, p<0.05) than HW. CPCs were positively correlated with abdominal adiposity in OW, and negatively correlated in HW with a significant difference between correlations (p<0.05). CPC content was positively correlated with executive processes in OW, and negatively correlated in HW with a significant difference in the strength of the correlations between groups (p<0.05 for correlation between OW and HW). Finally, CPC-CM from OW trended to increase neuroblast viability in vitro relative to HW (1.79 fold, p=0.07). These novel findings indicate that increased content of CPCs among OW children may play a role in preventing decrements in cognitive function via paracrine mechanisms.


Sujet(s)
Dysfonctionnement cognitif , Fonction exécutive/physiologie , Cellules souches hématopoïétiques , Inflammation , Obésité pédiatrique , Enfant , Dysfonctionnement cognitif/sang , Dysfonctionnement cognitif/immunologie , Dysfonctionnement cognitif/physiopathologie , Humains , Inflammation/sang , Inflammation/immunologie , Inflammation/physiopathologie , Mâle , Obésité pédiatrique/sang , Obésité pédiatrique/immunologie , Obésité pédiatrique/physiopathologie
12.
Cancer Cell Int ; 15: 42, 2015.
Article de Anglais | MEDLINE | ID: mdl-26060426

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) in tumors have emerged as progenitors involved in stroma formation and metastasis of cancers, partially owing to their abilities to differentially express paracrine factors related to the proliferation and invasion of cancer cells. In this regard, increasing evidence has shown that MSCs have impacts on the malignancy of colon cancer, however, the underpinning mechanisms by which MSCs promote cancer metastasis remain elusive. METHODS: To investigate the crosstalk between adipose-derived MSCs (AMSCs) isolated from adipose tissues and colon cancer cells, a co-culture transwell model of AMSCs and colon cancer cells was employed, and the activation of Wnt signaling and paracrine factors in colon cancer cells and AMSCs were measured. RESULTS: The results showed that AMSCs could enhance the metastatic capacity of colon cancer cells with an elevated expression of mesenchymal-epithelial transition (EMT)-associated genes in a contact-dependent manner. Reciprocally, colon cancer cells were able to induce AMSCs to produce metastasis-related factors and cytokines, such as FGF10, VEGFC and matrix metalloproteinases (MMPs) in part through a mechanism of an activation of Wnt signaling, by which these factors in turn activate Wnt signaling of colon cancer cells. Intriguingly, an inhibition of Wnt signaling leads a reduced capacity of invasion and colony formation of colon cancer cells in vitro, and the tumorigenicity of cancer cells in a murine model. CONCLUSIONS: These findings thus suggest that the crosstalk between the Wnt signaling of cancer cells and paracrine factors of AMSCs has an implication in colon cancer malignancy. This study thus uncovers a novel Wnt-paracrine factors mediated-crosstalk between colon cancer cells and AMSCs in cancer malignancy.

13.
Biomaterials ; 52: 318-26, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25818438

RÉSUMÉ

Stem cell therapies for atherosclerotic diseases are promising, but benefits remain modest with present cell delivery devices in part due to cell washout and immune attack. Many stem cell effects are believed mediated by paracrine factors (PFs) secreted by the stem cells which potentiate tissue repair via activation and enhancement of intrinsic host repair mechanisms We therefore sought to create an "intravascular paracrine factor factory" by harnessing stem cells on a stent using a nanofiber (NF) stent sleeve, and thus providing a sheltered milieu for cells to continuously produce PFs on-stent. The NF sleeve acts as a substrate on which stem cells grow, and as a semi-permeable barrier that protects cells from washout and host immune response while allowing free outward passage of PFs. NF stent sleeves were created by covering stents with electrospun poly-lactic-co-glycolic acid nanofibers and were then uniformly coated with mesenchymal stem cells (MSCs). NF sleeves blocked cell passage but did not hamper MSC attachment or proliferation, and did not alter MSC morphology or surface markers. NF sleeve MSCs continued to secrete PFs that were biologically active and successfully induced tubulogenesis in human endothelial cells. NF stent sleeves seeded with allogeneic MSCs implanted in pigs remained patent at 7 days without thrombotic occlusion or immune rejection. Our results demonstrate the feasibility of creating an intravascular PF factory using a stem cell impregnated NF stent sleeve, and pave the way for animal studies to assess the efficacy of local PF production to treat ischemic artery disease.


Sujet(s)
Systèmes de délivrance de médicaments , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Nanofibres/composition chimique , Endoprothèses , Angiographie , Animaux , Adhérence cellulaire , Prolifération cellulaire , Survie cellulaire , Matériaux revêtus, biocompatibles/composition chimique , Conception d'appareillage , Cellules endothéliales de la veine ombilicale humaine/cytologie , Humains , Ischémie/anatomopathologie , Perméabilité , Suidae , Transplantation homologue
14.
Circ Heart Fail ; 8(1): 167-74, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25342737

RÉSUMÉ

BACKGROUND: Stem cell factor (SCF), a ligand of the c-kit receptor, is a critical cytokine, which contributes to cell migration, proliferation, and survival. It has been shown that SCF expression increases after myocardial infarction (MI) and may be involved in cardiac repair. The aim of this study was to determine whether gene transfer of membrane-bound human SCF improves cardiac function in a large animal model of MI. METHODS AND RESULTS: A transmural MI was created by implanting an embolic coil in the left anterior descending artery in Yorkshire pigs. One week after the MI, the pigs received direct intramyocardial injections of either a recombinant adenovirus encoding for SCF (Ad.SCF, n=9) or ß-gal (Ad.ß-gal, n=6) into the infarct border area. At 3 months post-MI, ejection fraction increased by 12% relative to baseline after Ad.SCF therapy, whereas it decreased by 4.2% (P=0.004) in pigs treated with Ad.ß-gal. Preload-recruitable stroke work was significantly higher in pigs after SCF treatment (Ad.SCF, 55.5±11.6 mm Hg versus Ad.ß-gal, 31.6±12.6 mm Hg, P=0.005), indicating enhanced cardiac function. Histological analyses confirmed the recruitment of c-kit(+) cells as well as a reduced degree of apoptosis 1 week after Ad.SCF injection. In addition, increased capillary density compared with pigs treated with Ad.ß-gal was found at 3 months and suggests an angiogenic role of SCF. CONCLUSIONS: Local overexpression of SCF post-MI induces the recruitment of c-kit(+) cells at the infarct border area acutely. In the chronic stages, SCF gene transfer was associated with improved cardiac function in a preclinical model of ischemic cardiomyopathy.


Sujet(s)
Thérapie génétique/méthodes , Infarctus du myocarde/thérapie , Myocarde/métabolisme , Facteur de croissance des cellules souches/métabolisme , Débit systolique , Fonction ventriculaire gauche/physiologie , Animaux , Modèles animaux de maladie humaine , Femelle , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/physiopathologie , Myocarde/anatomopathologie , Suidae
15.
Vascul Pharmacol ; 63(1): 19-28, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24998908

RÉSUMÉ

The therapeutic effects of stem cell transplantation in ischemic disease are mediated by the production of paracrine bioactive factors. However, the bioactive factors secreted by human mesenchymal stem cells (hMSCs) and their angiogenic activity are not clearly identified or determined. We here found that hMSC-derived conditioned media (hMSC-CdM) stimulated in vitro angiogenic activity of endothelial cells and contained significant levels of various growth factors and cytokines, such as vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), monocyte chemotactic protein-1 (MCP-1), interleukin-6 (IL-6), and transforming growth factor-beta1 (TGF-ß1). The angiogenic activity of hMSC-CdM was significantly inhibited by pretreatment with neutralizing antibodies against VEGF, MCP-1, and IL-6, but not against TGF-ß1 and HGF. A mixture of those inhibitory antibodies blocked CdM-mediated activation of angiogenic signals, as well as inhibited CdM-mediated in vivo angiogenesis. Moreover, local injection of CdM increased angiogenesis and promoted blood flow in mice with hindlimb ischemia, and these effects were inhibited by co-treatment with these inhibitory antibodies. These results indicate that hMSC-CdM represents a promising cell-free therapeutic strategy for neovascularization in ischemic diseases. These results suggest the combination of VEGF, MCP-1, and IL-6 as a commercial application for therapeutic angiogenesis.


Sujet(s)
Ischémie/thérapie , Cellules souches mésenchymateuses/métabolisme , Néovascularisation physiologique/physiologie , Communication paracrine/physiologie , Adulte , Animaux , Chimiokine CCL2/métabolisme , Cytokines/métabolisme , Membre pelvien/vascularisation , Humains , Protéines et peptides de signalisation intercellulaire/métabolisme , Interleukine-6/métabolisme , Mâle , Souris , Souris de lignée C57BL , Adulte d'âge moyen , Facteur de croissance endothéliale vasculaire de type A/métabolisme
16.
Biomed Khim ; 60(2): 246-57, 2014.
Article de Russe | MEDLINE | ID: mdl-24837313

RÉSUMÉ

Adenosine, endogenous purine nucleoside, is an ATP metabolite that also acts as an extracellular signaling molecule. The concentration of extracellular adenosine rises during hypoxia and cell damage leading to numerous pleiotropic effects. Although a high concentration of adenosine was found at burn injury, the effect has not been well elucidated. We have studied human peripheral blood myeloid cell, due to their expression of specific adenosine receptors and capacity to migrate to the site of burn injury. We have shown that myeloid cells after 72 hours of stimulation of adenosine receptors develop altered expression of surface antigens: preserved monocyte's marker CD14 with already expressed dendritic cell markers (CD209, CD1a). Whereas untreated cells have already lost monocyte marker in 72 hours, and express CD1a more abundantly. Adenosine modified myeloid cells express also higher levels of mRNA of proinflammatory cytokines and chemoattractants (IL-6, IL-8, IL-1 b). Using mouse model of the burn injury we have shown, that adenosine modified bone marrow derived myeloid cells injected in the site of the injury promote migration of granulocytes, monocytes, macrophages, and fibroblasts on the 7th day after burn. Thus, stimulation of adenosine receptors alters differentiation and function of myeloid cells. In the site of burn injury adenosine modified myeloid cells augment cell migration due to paracrine factors.


Sujet(s)
Adénosine/métabolisme , Brûlures/immunologie , Chimiotaxie des leucocytes/immunologie , Monocytes/immunologie , Cellules myéloïdes/immunologie , Récepteurs purinergiques P1/métabolisme , Adénosine-5'-(N-éthylcarboxamide)/pharmacologie , Animaux , Antigènes CD/biosynthèse , Antigènes CD/immunologie , Brûlures/métabolisme , Différenciation cellulaire/immunologie , Cellules cultivées , Chimiotaxie des leucocytes/effets des médicaments et des substances chimiques , Cytokines/biosynthèse , Cytokines/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Modèles animaux de maladie humaine , Cytométrie en flux , Humains , Souris , Souris de lignée C57BL , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Cellules myéloïdes/effets des médicaments et des substances chimiques , Cellules myéloïdes/métabolisme , ARN messager/biosynthèse , Réaction de polymérisation en chaine en temps réel
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE