Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
APL Bioeng ; 8(2): 021501, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38572313

ABSTRACT

Cancer, with high morbidity and high mortality, is one of the major burdens threatening human health globally. Intervention procedures via percutaneous puncture have been widely used by physicians due to its minimally invasive surgical approach. However, traditional manual puncture intervention depends on personal experience and faces challenges in terms of precisely puncture, learning-curve, safety and efficacy. The development of puncture interventional surgery robotic (PISR) systems could alleviate the aforementioned problems to a certain extent. This paper attempts to review the current status and prospective of PISR systems for thoracic and abdominal application. In this review, the key technologies related to the robotics, including spatial registration, positioning navigation, puncture guidance feedback, respiratory motion compensation, and motion control, are discussed in detail.

2.
Small ; 20(24): e2307628, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38191883

ABSTRACT

Injectable bioadhesives are attractive for managing gastric ulcers through minimally invasive procedures. However, the formidable challenge is to develop bioadhesives that exhibit high injectability, rapidly adhere to lesion tissues with fast gelation, provide reliable protection in the harsh gastric environment, and simultaneously ensure stringent standards of biocompatibility. Here, a natural bioadhesive with tunable cohesion is developed based on the facile and controllable gelation between silk fibroin and tannic acid. By incorporating a hydrogen bond disruptor (urea or guanidine hydrochloride), the inherent network within the bioadhesive is disturbed, inducing a transition to a fluidic state for smooth injection (injection force <5 N). Upon injection, the fluidic bioadhesive thoroughly wets tissues, while the rapid diffusion of the disruptor triggers instantaneous in situ gelation. This orchestrated process fosters the formed bioadhesive with durable wet tissue affinity and mechanical properties that harmonize with gastric tissues, thereby bestowing long-lasting protection for ulcer healing, as evidenced through in vitro and in vivo verification. Moreover, it can be conveniently stored (≥3 m) postdehydration. This work presents a promising strategy for designing highly injectable bioadhesives utilizing natural feedstocks, avoiding any safety risks associated with synthetic materials or nonphysiological gelation conditions, and offering the potential for minimally invasive application.


Subject(s)
Hydrogen Bonding , Stomach Ulcer , Animals , Stomach Ulcer/drug therapy , Injections , Tissue Adhesives/chemistry , Adhesives/chemistry , Fibroins/chemistry , Tannins/chemistry , Rats, Sprague-Dawley
3.
Aesthetic Plast Surg ; 48(3): 519-529, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38148357

ABSTRACT

BACKGROUND: The fat retention rate is associated with postoperative inflammation. However, fat survival is still unpredictable even when supplemented with adipose-derived stem cells (ADSCs). Beige adipocytes play a role in regulating pathological inflammation. Thus, we assumed that exosomes may promote macrophage polarization to regulate inflammation when we simulated postgrafted inflammation by lipopolysaccharide (LPS) induction. METHODS: 3T3-L1 preadipocytes were used to differentiate into beige adipocytes, which were stimulated by special culture media, and then, exosomes were isolated from the supernatant. We identified them by morphology, protein and gene expression, or size distribution. Next, we utilized exosomes to stimulate LPS-induced macrophages and evaluated the changes in inflammatory cytokines and macrophage polarization. RESULTS: The induced cells contained multilocular lipid droplets and expressed uncoupling protein 1 (UCP1) and beige adipocyte-specific gene. The exosomes, which were approximately 111.5 nm and cup-like, were positive for surface markers. Additionally, the levels of proinflammatory-related indicators in the LPS+exosomes (LPS+Exos) group were increased after inflammation was activated for 6 h. When inflammation lasted 16 h, exosomes decreased the expression of proinflammatory-related indicators and increased the expression of anti-inflammatory-related indicators compared with the group without exosomes. CONCLUSION: The method described in this article can successfully obtain beige adipocytes and exosomes. The results suggest that beige adipocyte exosomes can promote inflammatory infiltration and polarize more macrophages to the M1 type in the early period of inflammation, accelerating the occurrence of the inflammation endpoint and the progression of macrophage switching from M1 to M2, while inflammation develops continuously. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Subject(s)
Adipocytes, Beige , Exosomes , Animals , Lipopolysaccharides/pharmacology , Macrophages , Inflammation
4.
BMC Cancer ; 23(1): 942, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37798663

ABSTRACT

BACKGROUND: Tumor necrosis factor receptor-associated factor 3 (TRAF3) has specific regulatory effects on a wide range of diseases, including tumors. However, the effect and mechanism of TRAF3 on lung adenocarcinoma (LUAD) are still unknown. The aim of the present study was to make clear the role and potential mechanism of TRAF3 in LUAD. METHODS: TIMER2.0 database and western blot were applied to detect the expression of TRAF3 in lung adenocarcinoma tissue. Kaplan-Meier Plotter database was utilized to explore the effect of TRAF3 on the clinical prognosis of lung adenocarcinoma patients. Specific siRNA was used to inhibit the expression of TRAF3 in LUAD cells (A549 and H1299). CCK-8 and EdU assays were performed for assessing LUAD cells proliferation. Wound healing assay and transwell assay were performed for determining cells migration. CCK-8 assay was used to assess the response of the LUAD cells to paclitaxel. TIMER2.0 bioinformatics and western blot were employed to detect the effects of TRAF3 on pyroptosis in LUAD. RESULTS: TRAF3 was highly expressed in lung adenocarcinoma tissues and cell lines. Patients with TRAF3 hyperexpression had a good prognosis compared to those with lower expression. TRAF3 inhibition notably induced proliferation and migration of LUAD cells. Inhibition of TRAF3 also weakened the sensitivity of LUAD cells to paclitaxel. Moreover, bioinformatics results showed that TRAF3 was positively correlated with the expression of pyroptosis-related genes in LUAD. Western blot assays showed that TRAF3 inhibition visibly decreased the expression of apoptosis-associated speck-like protein (ASC), cleaved caspase-1 and matured- IL-1ß. CONCLUSIONS: Inhibition of TRAF3 promotes the proliferation and migration of LUAD cells, and reduces the sensitivity of LUAD cells to paclitaxel. The effects of TRAF3 on LUAD cells were mediated in part by caspase-1-dependent pyroptosis.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Humans , Lung Neoplasms/pathology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Pyroptosis , Sincalide , Cell Line, Tumor , Adenocarcinoma of Lung/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Paclitaxel , Caspases/metabolism , Gene Expression Regulation, Neoplastic
5.
Respir Res ; 23(1): 151, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35681240

ABSTRACT

Small extracellular vesicles (sEVs) have been recognized to be more effective than direct stem cell differentiation into functional target cells in preventing tissue injury and promoting tissue repair. Our previous study demonstrated the protective effect of adipose-derived stem cells (ADSCs) on lipopolysaccharide (LPS)-induced acute lung injury and the effect of autophagy on ADSC functions, but the role of ADSC-derived sEVs (ADSC-sEVs) and autophagy-mediated regulation of ADSC-sEVs in LPS-induced pulmonary microvascular barrier damage remains unclear. After treatment with sEVs from ADSCs with or without autophagy inhibition, LPS-induced human pulmonary microvascular endothelial cell (HPMVECs) barrier damage was detected. LPS-induced acute lung injury in mice was assessed in vivo after intravenous administration of sEVs from ADSCs with or without autophagy inhibition. The effects of autophagy on the bioactive miRNA components of ADSC-sEVs were assessed after prior inhibition of cell autophagy. We found that ADSC-sEV effectively alleviated LPS-induced apoptosis, tight junction damage and high permeability of PMVECs. Moreover, in vivo administration of ADSC-sEV markedly inhibited LPS-triggered lung injury. However, autophagy inhibition, markedly weakened the therapeutic effect of ADSC-sEVs on LPS-induced PMVECs barrier damage and acute lung injury. In addition, autophagy inhibition, prohibited the expression of five specific miRNAs in ADSC-sEVs -under LPS-induced inflammatory conditions. Our results indicate that ADSC-sEVs protect against LPS-induced pulmonary microvascular barrier damage and acute lung injury. Autophagy is a positive mediator of sEVs function, at least in part through controlling the expression of bioactive miRNAs in sEVs.


Subject(s)
Acute Lung Injury , Extracellular Vesicles , MicroRNAs , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Autophagy/physiology , Extracellular Vesicles/metabolism , Lipopolysaccharides/toxicity , Mice , MicroRNAs/metabolism
6.
Cell Death Dis ; 10(11): 804, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31645547

ABSTRACT

Adipose-derived stem cells (ADSCs) have been shown to be beneficial in some pulmonary diseases, and the paracrine effect is the major mechanism underlying ADSC-based therapy. Autophagy plays a crucial role in maintaining stem cell homeostasis and survival. However, the role of autophagy in mediating ADSC paracrine effects has not been thoroughly elucidated. We examined whether ADSCs participate in lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial cell (PMVEC) barrier damage in a paracrine manner and illuminated the role of autophagy in regulating ADSC paracrine effects. PMVECs and ADSCs with or without autophagy inhibition were cocultured without intercellular contact, and the microvascular barrier function was assessed after LPS treatment. ADSC paracrine function was evaluated by detecting essential growth factors for endothelial cells. For in vivo experiments, ADSCs with or without autophagy inhibition were transplanted into LPS-induced lung-injury mice, and lung injury was assessed. ADSCs significantly alleviated LPS-induced microvascular barrier injury. In addition, ADSC paracrine levels of VEGF, FGF, and EGF were induced by LPS treatment, especially in the coculture condition. Inhibiting autophagy weakened the paracrine function and the protective effects of ADSCs on microvascular barrier injury. Moreover, ADSC transplantation alleviated LPS-induced lung injury, and inhibiting autophagy markedly weakened the therapeutic effect of ADSCs on lung injury. Together, these findings show that ADSC paracrine effects play a vital protective role in LPS-induced pulmonary microvascular barrier injury. Autophagy is a positive mediating factor in the paracrine process. These results are helpful for illuminating the role and mechanism of ADSC paracrine effects and developing effective therapies in acute lung injury.


Subject(s)
Acute Lung Injury/pathology , Lipopolysaccharides/metabolism , Stem Cells/metabolism , Adipocytes , Animals , Autophagy , Cell Differentiation , Mice
7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(12): 1498-1510, 2018 12.
Article in English | MEDLINE | ID: mdl-30296594

ABSTRACT

Some studies have shown that transplanted fat tissues usually cannot survive for long if adipose-derived stem cells (ADSCs) are removed from the tissues in advance. It is more meaningful to explore the mechanism mediating survival and differentiation of ADSCs in the transplanted microenvironment. AMP-activated protein kinase (AMPK) has been shown to be one of the energy receptors that regulate many aspects of cellular metabolism. AMPK activation has been implicated in models of adult ischemic injury, but the mechanism and the regulating effects of AMPK on survival and adipogenesis of transplanted ADSCs are still little known. In this study, we simulated the transplanted microenvironment using oxygen-glucose deprivation (OGD) to test the survival and adipogenesis of ADSCs. We found that OGD treatment triggered significant apoptosis and promoted autophagy. Simultaneously, OGD hindered the differentiation of ADSCs into mature adipocytes. After inhibiting AMPK, the OGD-induced apoptosis rate increased but autophagy was inhibited. The adipogenesis level also decreased. To show that the effects of AMPK on apoptosis and adipogenesis were autophagy-dependent, we pre-inhibited or pre-promoted autophagy with siATG7 or rapamycin while blocking AMPK. We found that inhibiting or improving autophagy exacerbated or alleviated the role of AMPK prohibition in apoptosis and adipogenesis. Furthermore, we showed that AMPK inhibition significantly lowered ULK1 activity but promoted mTOR activity, so that to inhibit autophagy. Our study shows that AMPK plays a protective role in maintaining survival and adipogenesis of OGD-challenged ADSCs partly by positively regulating autophagy. AMPK positively regulates autophagy by inhibiting mTOR but promoting ULK1 activity in OGD condition.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipogenesis , Adipose Tissue/cytology , Autophagy , Adipose Tissue/metabolism , Autophagy/drug effects , Autophagy-Related Protein 7/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/metabolism , Cell Differentiation , Cell Hypoxia , Cell Survival , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins/metabolism , RNA, Small Interfering/pharmacology , Sirolimus/pharmacology , Stem Cells/cytology , Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism
8.
J Cell Physiol ; 233(1): 688-698, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28328069

ABSTRACT

Understanding the role and underlying regulation mechanism of autophagy in lipopolysaccharide-induced lung injury (LPS-LI) may provide potentially new pharmacological targets for treatment of acute lung injury. The aim of this study was to investigate the functional significance of autophagy in LPS-LI. The autophagy of human pulmonary microvascular endothelial cells (HPMVECs) and mice was inhibited before they were challenged with LPS. In vitro, permeability, vitality, and the LDH release rate of the cells were detected, the zonula occluden-1 (ZO-1) expression and the stress fiber formation were determined. In vivo, the lung injury was assessed. We found LPS caused high permeability and increased lactate dehydrogenase (LDH) release rate, lowered viability of the cells, inhibited the ZO-1 expression and induced stress fiber formation, these effects were further aggravated by prohibiting the level of autophagy. Consistently, in in vivo experiments, LPS-induced serious lung injury, which was reflected as edema, leukocyte infiltration and hemorrhage in lung tissue, and the high concentration of pro-inflammation cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-1ß in bronchoalveolar lavage fluid (BALF). Inhibiting autophagy further exacerbated LPS-LI. It appears that autophagy played a protective role in LPS-LI in part through restricting the injury of lung microvascular barrier.


Subject(s)
Acute Lung Injury/metabolism , Autophagy , Blood-Air Barrier/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Lipopolysaccharides , Lung/blood supply , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Blood-Air Barrier/ultrastructure , Cells, Cultured , Disease Models, Animal , Endothelial Cells/ultrastructure , Humans , Lung/metabolism , Lung/ultrastructure , Mice, Inbred C57BL , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , RNA Interference , Stress Fibers/metabolism , Transfection , Zonula Occludens-1 Protein/metabolism
9.
Cell Physiol Biochem ; 44(5): 1762-1774, 2017.
Article in English | MEDLINE | ID: mdl-29216630

ABSTRACT

BACKGROUND/AIMS: Ischemia is one of the main causes of the high rate of absorption of transplanted autologous fat. Autophagy allows cells to survive by providing energy under starvation. Rapamycin has been found to play a role in promoting autophagy. In this study, we investigated whether rapamycin participates in the survival and adipogenesis of ischemia-challenged adipose-derived stem cells (ADSCs) by regulating autophagy. METHODS: Before the cells were exposed to oxygen-glucose deprivation (OGD), a simulated ischemic microenvironment, the level of autophagy was reduced or increased by lentiviral transfection with short hairpin RNA targeting microtubule-associated protein 1-light chain 3 gene (shRNA-LC3) or treatment with rapamycin, respectively. The level of autophagy was assessed by western blotting, transmission electron microscopythen the apoptosis ratio was determined through terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) and flow cytometry. Adipogenesis was further evaluated by oil red O staining and the expressions level of some specific proteins for adipocytes. RESULTS: shRNA-LC3 and rapamycin treatment effectively decreased and improved the level of autophagy in cells with or without OGD challenge, respectively. In addition, autophagy inhibition increased the apoptosis rate and activated caspase-3 expression level in response to OGD, and these were markedly inhibited by rapamycin preconditioning. During adipogenesis, autophagy inhibition decreased not only oil droplet accumulation but also lipoprotein lipase (LPL) and peroxisome proliferator-activated receptor gamma (PPARγ) expression in cells with or without OGD challenge. However, autophagy promotion by rapamycin increased oil droplet accumulation and LPL and PPARγ expression. CONCLUSIONS: Rapamycin may promote the survival and adipogenesis of ischemia-challenged ADSCs by upregulating autophagy.


Subject(s)
Adipogenesis/drug effects , Autophagy/drug effects , Microtubule-Associated Proteins/metabolism , Sirolimus/pharmacology , Adipose Tissue/cytology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Hypoxia , Cell Line , Cell Survival/drug effects , Glucose/metabolism , Humans , Ischemia/metabolism , Ischemia/pathology , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , PPAR gamma/metabolism , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/ultrastructure , Transfection
10.
Am J Physiol Lung Cell Mol Physiol ; 313(2): L384-L394, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28522565

ABSTRACT

Integrin αvß5 mediates pulmonary endothelial barrier function and acute lung injury (LI), but its roles in cell apoptosis and autophagy are unclear. Thus, the aims of this study were to investigate the significance of αvß5 in ischemia-reperfusion (I/R)-induced apoptosis and LI and to explore the relationship between αvß5 and autophagy. Human pulmonary microvascular endothelial cells (HPMVECs) were pretreated with an αvß5-blocking antibody (ALULA) and challenged with oxygen-glucose deprivation/oxygen-glucose restoration, which mimics I/R; then, cellular autophagy and apoptosis were detected, and cell permeability was assessed. In vivo, mice were pretreated with the autophagy inhibitor chloroquine (CLQ), followed by treatment with ALULA. The mice then underwent operative lung I/R. LI was assessed by performing a pathological examination, calculating the wet/dry lung weight ratio and detecting the bronchial alveolar lavage fluid (BALF) protein concentration. αvß5 inhibition promoted HPMVEC autophagy under I/R in vitro, alleviated cell permeability, decreased the apoptosis ratio, and activated caspase-3 expression. These outcomes were significantly diminished when autophagy was inhibited with a small-interfering RNA construct targeting autophagy-related gene 7 (siATG7). Moreover, ALULA pretreatment alleviated I/R-induced LI (I/R-LI), which manifested as a decreased wet/dry lung weight ratio, an altered BALF protein concentration, and lung edema. Preinhibiting autophagy with CLQ, however, eliminated the protective effects of ALULA on I/R-LI. Therefore, inhibiting αvß5 effectively ameliorated I/R-induced endothelial cell apoptosis and I/R-LI. This process was dependent on improved autophagy and its inhibitory effects on activated caspase-3.


Subject(s)
Acute Lung Injury/drug therapy , Autophagy/drug effects , Lung/drug effects , Protective Agents/pharmacology , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/metabolism , Reperfusion Injury/drug therapy , Acute Lung Injury/metabolism , Animals , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Caspase 3/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glucose/metabolism , Humans , Lung/metabolism , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Reperfusion Injury/metabolism
11.
Sci Rep ; 7: 45360, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28350007

ABSTRACT

Fabricating Ti alloy based dental implants with defined porous scaffold structure is a promising strategy for improving the osteoinduction of implants. In this study, we use Laser Beam Melting (LBM) 3D printing technique to fabricate porous Ti6Al4V dental implant prototypes with three controlled pore sizes (200, 350 and 500 µm). The mechanical stress distribution in the surrounding bone tissue is characterized by photoelastography and associated finite element simulation. For in-vitro studies, experiments on implants' biocompatibility and osteogenic capability are conducted to evaluate the cellular response correlated to the porous structure. As the preliminary results, porous structured implants show a lower stress-shielding to the surrounding bone at the implant neck and a more densed distribution at the bottom site compared to the reference implant. From the cell proliferation tests and the immunofluorescence images, 350 and 500 µm pore sized implants demonstrate a better biocompatibility in terms of cell growth, migration and adhesion. Osteogenic genes expression of the 350 µm group is significantly increased alone with the ALP activity test. All these suggest that a pore size of 350 µm provides an optimal provides an optimal potential for improving the mechanical shielding to the surrounding bones and osteoinduction of the implant itself.


Subject(s)
Biocompatible Materials/chemistry , Dental Implants , Titanium/chemistry , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Alloys , Animals , Biocompatible Materials/pharmacology , Cell Line , Cell Survival/drug effects , Elasticity , Lasers , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Porosity , Printing, Three-Dimensional
12.
Int J Clin Exp Pathol ; 10(9): 9310-9317, 2017.
Article in English | MEDLINE | ID: mdl-31966803

ABSTRACT

Inhibiting inflammation is helpful in relieving the absorption of alveolar bone and promoting periodontal bone regeneration. In a previous study, we showed that transforming growth factor-beta1 (TGF-ß1)-induced Treg cells inhibit the absorption of tissue-engineered cartilage caused by endogenous IFN-γ and TNF-α. In this study, we investigated the effect of inhibiting local inflammatory responses on Geistlich Bio-Oss® osteogenesis promotion in vivo. TGF-ß1+BMMSCs (bone marrow mesenchymal stem cells) were cultured in Geistlich Bio-Oss® medium, and biocompatibility was evaluated. Alveolar bone defects in New Zealand rabbits repaired by application of Geistlich Bio-Oss® were compared to the effects of added TGF-ß1+BMMSCs. There was no significant difference between the untreated Geistlich Bio-Oss® medium-control group and the group treated with the addition of TGF-ß1+BMMSCs. Pro-inflammatory cytokines IFN-γ and TNF-α delayed Geistlich Bio-Oss®-induced osteogenesis, but no significant difference in osteogenesis was seen with the addition of TGF-ß1+BMMSCs. Geistlich Bio-Oss® has good compatibility with TGF-ß1+BMMSCs. However, the dual role of in vivo TGF-ß1+BMMSCs in regenerating periodontal bone and limiting local inflammation is not clear.

13.
J Tissue Eng Regen Med ; 10(2): E113-20, 2016 Feb.
Article in English | MEDLINE | ID: mdl-23868873

ABSTRACT

The objective of the present study was to explore the mechanisms of transforming growth factor (TGF)-ß1 inhibiting the absorption of tissue engineering cartilage. We transfected TGF-ß1 gene into bone marrow mesenchymal stem cells (BMMSCs) and co-cultured with interferon (IFN)-γ and tumour necrosis factor (TNF)-α and CD4(+) CD25(-) T lymphocytes. We then characterized the morphological changes, apoptosis and characterization of chondrogenic-committed cells from TGF-ß1(+) BMMSCs and explored their mechanisms. Results showed that BMMSCs apoptosis and tissue engineering cartilage absorption in the group with added IFN-γ and TNF-α were greater than in the control group. In contrast, there was little BMMSC apoptosis and absorption by tissue engineering cartilage in the group with added CD4(+) CD25(-) T lymphocytes; Foxp3(+) T cells and CD25(+) CD39(+) T cells were found. In contrast, no type II collagen or Foxp3(+) T cells or CD25(+) CD39(+) T cells was found in the TGF-ß1(-) BMMSC group. The data suggest that IFN-γ and TNF-α induced BMMSCs apoptosis and absorption of tissue engineering cartilage, but the newborn regulatory T (Treg) cells inhibited the function of IFN-γ and TNF-α and protected BMMSCs and tissue engineering cartilage. TGF-ß1not only played a cartilage inductive role, but also inhibited the absorption of tissue engineering cartilage. The pathway proposed in our study may simulate the actual reaction procedure after implantation of BMMSCs and tissue engineering cartilage in vivo.


Subject(s)
Absorption, Physiological , Cartilage/metabolism , T-Lymphocytes, Regulatory/metabolism , Tissue Engineering/methods , Transforming Growth Factor beta1/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Cell Lineage , Cell Shape , Chondrogenesis , DNA Fragmentation , DNA, Complementary/genetics , Female , In Situ Nick-End Labeling , Interferon-gamma/metabolism , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Transforming Growth Factor beta1/biosynthesis
14.
Mol Med Rep ; 12(3): 4230-4237, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26096280

ABSTRACT

Bone mesenchymal stem cells (BMSCs) have been an area of interest in biomedical research and tissue engineering due to their diverse differentiation abilities. In osteogenesis, bone morphogenetic proteins (BMPs), particularly BMP­2, are important. However, the effect of BMP­2 on the osteogenetic capacity of BMSCs remains to be fully elucidated. In the present study, primary rat BMSCs were infected with a recombinant lentivirus carrying the BMP­2 gene (Lenti­BMP­2), and the effects of BMP­2 on the activity of alkaline phosphatase (ALP) on days 3, 7, 14 and 21, and on mineralization on day 21 were evaluated. In addition, the adhesive ability of BMP­2­overexpressed BMSCs was detected using an adhesion assay. Following forced expression of BMP­2 in the BMSCs, the levels of osteogenic genes, including osteopontin (OPN), osteocalcin (OC) and collagen type I (Col­â… ), were detected and the nuclear accumulation of Runt­related transcription factor (Runx)­2 and phosphorylated small mothers against decapentaplegic (p­Smad) 1/5/8 was also evaluated. The results demonstrated that the rat BMSCs had been isolated, cultured and passaged from Sprague­Dawley rat bone marrow successfully, and the third­generation BMSCs were identified using flow cytometry with CD29 staining. The osteogenetic phenotype of the BMSCs, expressing ALP and osteocalcin, was significantly induced by BMP­2, and the proliferation of the BMSCs was enhanced by BMP­2. Furthermore, the adhesive potential of the BMP­2­overexpressed BMSCs was increased, the expression levels of OPN, OCN and Col­â… e osteogenetic factors were upregulated and the nuclear accumulation of Runx­2 and p­Smads1/5/8 were increased significantly. These data suggested that BMP­2 may facilitate the osteogenetic differentiation of rat BMSCs and provide a favorable cell resource for tissue engineering.


Subject(s)
Bone Marrow Cells/cytology , Bone Morphogenetic Protein 2/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Cell Adhesion , Cell Differentiation , Cells, Cultured , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/cytology , Osteocalcin/metabolism , Osteogenesis , Osteopontin/metabolism , Rats , Rats, Sprague-Dawley , Smad Proteins/metabolism
15.
J Heart Lung Transplant ; 34(5): 746-55, 2015 May.
Article in English | MEDLINE | ID: mdl-25934479

ABSTRACT

BACKGROUND: Understanding the role and underlying regulation mechanism of autophagy in ischemia/reperfusion (I/R)-induced lung injury may provide potentially new pharmacologic targets for treatment of acute lung injury. The aim of this study was to adjust autophagy with pharmacologic agents to determine its functional significance in I/R-induced lung injury. METHODS: Human pulmonary microvascular endothelial cells (HPMVECs) and mice were pre-conditioned with autophagy inhibitor chloroquine or promoter rapamycin before they were challenged with oxygen-glucose deprivation/oxygen-glucose restoration (OGD) and lung I/R, respectively. Extracellular signal-regulated kinase (ERK)1/2 inhibitor U0126 was pre-injected into I/R-induced mice to test the role of ERK1/2 in regulating autophagy. RESULTS: OGD caused tight conjunction damage and cell death in HPMVECs, which was further aggravated by blocking autophagy, yet ameliorated through promoting autophagy. On a consistent basis, inhibiting autophagy aggravated I/R-induced lung edema and tissue inflammation, which was significantly alleviated by promoting autophagy with rapamycin. In addition, inhibition of ERK1/2 increased expression of active mammalian target-of-rapamycin and thus decreased I/R-induced autophagy. CONCLUSIONS: It appears that autophagy plays a protective role in I/R-induced lung injury and this effect may be enhanced by moderately improving autophagy level. Meanwhile, the ERK1/2 signal pathway has a positively regulating role in lung I/R-induced autophagy.


Subject(s)
Autophagy/physiology , Blood-Air Barrier/physiology , Endothelium, Vascular/metabolism , Lung Injury/pathology , Reperfusion Injury/pathology , Animals , Cell Survival , Cells, Cultured , Endothelium, Vascular/pathology , Humans , Lung Injury/metabolism , Mice , Reperfusion Injury/metabolism
16.
Expert Opin Biol Ther ; 14(5): 573-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24617422

ABSTRACT

BACKGROUND: In a previous study, we showed that IFN-γ and TNF-α induce bone marrow mesenchymal stem cell (BMMSC) apoptosis and absorption of tissue-engineered cartilage, but the induced regulatory T cells (iTreg) inhibit the function of IFN-γ and TNF-α. In this study, we investigated the effect of iTreg cells on the absorption of tissue-engineered cartilage caused by endogenous IFN-γ and TNF-α. METHODS: We transfected the TGF-ß1 gene into BMMSCs co-cultured with CD4(+) T lymphocytes. Then, we assessed the expression of iTreg cell markers (Foxp3, CD25 and CD 39) and IFN-γ and TNF-α and the level of apoptosis of BMMSCs. In addition, we characterized chondrogenic-committed cells from TGF-ß1(+)BMMSCs and explored the role of iTreg cells. RESULTS: IFN-γ and TNF-α were detected in the groups with CD4(+) T cells. In the group in which TGF-ß1(+)BMMSCs were co-cultured with CD4(+) T cells, we observed Foxp3(+)Treg/ CD25(+)CD39(+) (17.58 ± 0.45%) cells as well as significant inhibition of BMMSC apoptosis and tissue-engineered cartilage absorption. CONCLUSIONS: CD4(+) T cells led to the absorption of tissue-engineered cartilage through the secretion of endogenous IFN-γ and TNF-α, whose inflammatory functions were concomitantly suppressed by iTreg cells converted from CD4(+) T cells. This study is clinically relevant and adds to our understanding of the mechanism of tissue-engineered cartilage absorption.


Subject(s)
Cartilage/metabolism , Interferon-gamma/metabolism , T-Lymphocytes, Regulatory/immunology , Tissue Engineering , Transforming Growth Factor beta1/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Base Sequence , Coculture Techniques , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics
17.
Expert Opin Biol Ther ; 12(9): 1129-39, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22762256

ABSTRACT

BACKGROUND: Skin and soft tissue expansion has limitations such as long hospitalization time and flap retraction after expansion. Our previous study suggested that bone marrow-derived stem cells contribute skin regeneration in skin and soft tissue expansion. In this study, the authors explored the feasibility of applying the bone marrow mesenchymal stem cells (BMMSCs) to the treatment of skin and soft tissue expansion and increasing the skin regeneration efficiency. METHODS: Sixty silicone expanders were implanted in the backs of 15 pigs, and allogeneic BMMSCs were transplanted to skin shallow fascia layer (local transplantation, Group A) or via ear vein (systemic transplantation, Group B). Group C was the Sham operation control; and then the expanders were injected with normal saline (N.S.). Skin was obtained at different time points (days 0, 14, 21, 28, 35, and 42). The organizational structure changes of the target skin were observed in the expansion process. The distribution, differentiation, and paracrine function of labeled BMMSCs were detected. RESULTS: Comparing with Group B (25.00 ± 1.98 cm(2)) or Group C (24.00 ± 1.10 cm(2), no transplantation), the expanded skin area of Group A (28.82 ± 1.43 cm(2)) increased, with the morphology of epidermis thickened, and dermis thinned. The BMMSCs differentiated into vascular endothelial cells and dermal fibroblasts. The quantity of newborn cells was proportional to the number of transplanted cells. The gene expression of VEGF, bFGF, EGF, and SDF in Group A was higher than those in Group B or C. The most obvious changes were on day 35. CONCLUSIONS: The local transplanted BMMSCs could increase the skin regeneration efficiency in skin and soft tissue expansion and reduce skin shrinkage effectively after removing the expander. Growth factors, VEGF, bFGF, EGF, and SDF, are favorable to this process.


Subject(s)
Bone Marrow Transplantation , Dermatologic Surgical Procedures/methods , Fascia/pathology , Fasciotomy , Mesenchymal Stem Cell Transplantation , Regeneration , Skin/pathology , Surgical Flaps , Animals , Cell Differentiation , Cell Proliferation , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Dermatologic Surgical Procedures/instrumentation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fascia/blood supply , Fascia/metabolism , Feasibility Studies , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Male , Mice , Phenotype , Prosthesis Design , Silicones , Skin/blood supply , Skin/metabolism , Swine , Time Factors , Tissue Expansion Devices , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
Zhonghua Zheng Xing Wai Ke Za Zhi ; 27(3): 207-12, 2011 May.
Article in Chinese | MEDLINE | ID: mdl-21838003

ABSTRACT

OBJECTIVE: To study the immuno-tolerance mechanism of the third-party bone marrow-derived mesenchymal stem cells (BMSCs) in the allogeneic transplantation. METHODS: Forty female C57BL/6 mice and forty male BALB/C mice were respectively used as donors and recipients in skin allogenic graft model. Forty male BALB/C mice were divided randomly into 4 groups: blank control group, CP group, BMSCs group , CP + BMSCs group, with 10 mice in each group. Before skin graft, high-dose abdominal injection of cyclophosphamide (200 mg/kg, 2 d, q. d.) was performed in recipient mice in CP and CP + BMSCs groups. On the transplantation day, a bonus of 2 x 10(6) BMSCs from the SD rat (SD-BMSCs) were injected through the tail vein in the BMSCs and CP + BMSCs groups. The observation and HE staining of skin grafts were used. The expressions of CD29, CD34, CD45 and CD90 of cells were analyzed by using flow cytometry in order to identify BMSCs. The CD4+, CD25+, Foxp3 and Treg cells of spleen were detected by flow cytometry. Cytokine in peripheral blood of recipient mice were measured by ELISA, including TGF-beta, IL-10 and IFN-gamma. T cells were co-cultured with 60Co-irradiated bone marrow MSCs from different individuals. The proliferative activity of T cells were evaluated with MTT assay. RESULTS: The skin graft survival time was significantly prolonged in the CP + BMSCs group, as compared with that in the blank control group, the CP group, the BMSCs group, respectively. Cells cultured by whole bone marrow adherent cultivation showed CD29 (99.7%), CD44+ (96.7%), CD34- (1.6%), CD45- (1.3%). Compared with the control group and CP group, the ratio of the CD4+, CD25+, Foxp3+ and Treg cells significantly increased in the SD-BMSCs group and CP + BMSCs group (P < 0.05). Analysis of peripheral blood by ELISA showed significant high level of TGF-beta, IL-10 and low level of IFN-gamma in BMSCs group and CP group,compared with that in control group. When co-cultured with BMSCs from different individuals, T- lymphocytes proliferation decreased apparently in SD-BMSCs group and C57-BMSCs group (P < 0.05), but there was no significant difference between SD-BMSCs group and C57-BMSCs group (P > 0.05). CONCLUSIONS: The immunotolerance mechanism of the third-party bone marrow-derived mesenchymal stem cells in the allogeneic transplantation might be associated with its effect on the proliferation of Treg cells and increasing expression of TGF-beta and IL-10, decreasing expression of IFN-gamma.


Subject(s)
Bone Marrow Cells/immunology , Immune Tolerance , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Skin Transplantation , Animals , Female , Interferon-gamma/immunology , Interleukin-10/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Transplantation, Homologous
19.
J Cell Physiol ; 226(11): 2834-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21302285

ABSTRACT

Skin and soft tissue expansion stimulates the proliferation of skin epidermal basal cells and increase the dermal collagen deposition and angiogenesis. To explore the contribution of bone marrow-derived stem cells (BMSCs) to the generation of "new" skin during the expansion, we used a chimeric mouse model in which the donor C57BL mice were engrafted with the bone marrow of enhanced green fluorescent protein (EGFP) transgenic mice. BMSCs were collected from the tibia and femur of EGFP(+) transgenic mice, and then injected into normal C57BL mice via the tail vein (chimeric mice). Skin was obtained at different times (days 0, 7, 14, 21, 28, and 35). Skin stromal-derived factor-1 (SDF-1) expression was evaluated. The number, distribution, and phenotype changes of EGFP(+) cells in the skin were also evaluated by means of fluorescent microscopy. EGFP(+) cells were present stably in the normal skin. The number of EGFP(+) cells of the Group A mice changed with the tension, and reached the peak on day 21(17.1 ± 6.7%), as compared with either Group B (5.5 ± 1.0%) or Group C (5.1 ± 0.9%). The SDF-1 expression in the expanded skin was significant increased (≈11-fold, P < 0.01) compared to non-expanded skin on day 21. Immunofluorescence showed EGFP(+) cells were converted into vascular endothelial cells, epidermal cells, and spindle-shaped dermal fibroblasts. Strain can promote the expression of SDF-1 and facilitate the differentiation and proliferation of BMSCs in the expanded skin.


Subject(s)
Bone Marrow Cells/cytology , Regeneration , Skin Physiological Phenomena , Stem Cells/physiology , Tissue Expansion , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Chemokine CXCL12/biosynthesis , Chimera/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostheses and Implants , Silicones/metabolism , Skin/cytology
SELECTION OF CITATIONS
SEARCH DETAIL