Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
ACS Biomater Sci Eng ; 10(5): 3316-3330, 2024 May 13.
Article En | MEDLINE | ID: mdl-38619014

In this study, we propose a spatially patterned 3D-printed nanohydroxyapatite (nHA)/beta-tricalcium phosphate (ß-TCP)/collagen composite scaffold incorporating human dental pulp-derived mesenchymal stem cells (hDP-MSCs) for bone regeneration in critical-sized defects. We investigated angiogenesis and osteogenesis in a rabbit critical-sized mandibular defect model treated with this engineered construct. The critical and synergistic role of collagen coating and incorporation of stem cells in the regeneration process was confirmed by including a cell-free uncoated 3D-printed nHA/ß-TCP scaffold, a stem cell-loaded 3D-printed nHA/ß-TCP scaffold, and a cell-free collagen-coated 3D-printed nHA/ß-TCP scaffold in the experimental design, in addition to an empty defect. Posteuthanasia evaluations through X-ray analysis, histological assessments, immunohistochemistry staining, histomorphometry, and reverse transcription-polymerase chain reaction (RT-PCR) suggest the formation of substantial woven and lamellar bone in the cell-loaded collagen-coated 3D-printed nHA/ß-TCP scaffolds. Histomorphometric analysis demonstrated a significant increase in osteoblasts, osteocytes, osteoclasts, bone area, and vascularization compared to that observed in the control group. Conversely, a significant decrease in fibroblasts/fibrocytes and connective tissue was observed in this group compared to that in the control group. RT-PCR indicated a significant upregulation in the expression of osteogenesis-related genes, including BMP2, ALPL, SOX9, Runx2, and SPP1. The findings suggest that the hDP-MSC-loaded 3D-printed nHA/ß-TCP/collagen composite scaffold is promising for bone regeneration in critical-sized defects.


Bone Regeneration , Calcium Phosphates , Ceramics , Hydrogels , Mandible , Neovascularization, Physiologic , Printing, Three-Dimensional , Tissue Scaffolds , Animals , Rabbits , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Humans , Ceramics/chemistry , Calcium Phosphates/chemistry , Hydrogels/chemistry , Osteogenesis/drug effects , Mesenchymal Stem Cells/metabolism , Collagen/chemistry , Durapatite/chemistry , Tissue Engineering/methods , Dental Pulp/cytology , Disease Models, Animal , Male , Angiogenesis
2.
Cell Tissue Bank ; 23(4): 923-936, 2022 Dec.
Article En | MEDLINE | ID: mdl-35590084

The aim of this research is to compare the capabilities of Adipose tissue mesenchymal stem cells (AT-MSCs) and bone marrow mesenchymal stem cells (BM-MSCs) in the treatment of diabetic male mice with CLI model. Supernatants were collected from C57BL/6 mice isolated AT-MSCs and BM-MSCs, afterward their effects on human umbilical vein endothelial (HUVEC) migration potential were evaluated. Diabetes mellitus type 1 was induced by streptozotocin injection. Diabetic mice with CLI model were divided into three groups and injected with AT-MSCs, BM-MSCs, or PBS then the efficacy of them was assessed. Survival of MSCs was analysed by SRY-specific gene. The conditioned medium of AT-MSCs and BM-MSCs stimulated HUVECs migration and the donor cells were detected till 21 day in two groups. BM-MSCs and AT-MSCs improved significantly functional recovery and ischemia damage. Neovascularization in ischemic muscle was significantly higher in mice treated with AT-MSCs and BM-MSCs and they improved muscle regeneration. In vivo and in vitro findings show that AT-MSCs and BM-MSCs transplantation could be proposed as a promising therapy to promote angiogenesis and muscle regeneration through secretion of proangiogenic factors, cytokines and growth factors in diabetic mice with CLI model wherein blood supply is insufficient and disrupted.


Diabetes Mellitus, Experimental , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mice , Male , Animals , Neovascularization, Physiologic , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/metabolism , Chronic Limb-Threatening Ischemia , Mice, Inbred C57BL , Mesenchymal Stem Cells/metabolism , Ischemia/therapy , Ischemia/metabolism , Adipose Tissue
3.
Microvasc Res ; 141: 104339, 2022 05.
Article En | MEDLINE | ID: mdl-35151721

PURPOSE: Critical limb ischemia (CLI) is the most severe manifestation of peripheral artery disease that diabetes mellitus is one of its major risk factors. MiR-126 as an endothelial cells specific miRNA plays a main role in angiogenesis. The objective of this study was to find a promising treatment by increasing therapeutic potential of adipose tissue mesenchymal stem cells (AT-MSCs) with microRNA-126 in diabetic mouse model with critical limb ischemia. AT-MSCs were isolated from male C57BL/6 mouse and characterized. METHODS: The cells were infected with miR-126 recombinant lentiviral vectors. Diabetes mellitus type 1 was induced and CLI was created in the animals. Animals were divided in different groups to receive PBS, MSCs, miR-126, and MSCmiR-126 and after the experiment, behavioural tests, cell survival, real-time PCR, and histopathological analysis were assessed. RESULTS: The results of function scores, VEGF-A level, and histopathology data demonstrated that the miR-126 treated group was better than PBS and MSCs groups. The expression of PIK3R2 and SPRED1 were decreased in miR-126 group compared to the control group. Our results showed that MSCsmiR-126 can live longer than MSCs in the gastrocnemius muscle. We conclude that mice treated with MSCsmiR-126 in functional tests showed better results and also the expression of VEGF-A and Microvessel density in them were higher than other groups. CONCLUSIONS: This study suggested that AT-MSCs overexpressing miR-126 could be an efficient therapeutic approach for angiogenesis in CLI with diabetes by downregulating SPRED1 and PIK3R2 and increasing secretion of angiogenic cytokines which can prolong the MSC survival.


Diabetes Mellitus , Mesenchymal Stem Cell Transplantation , MicroRNAs , Animals , Chronic Limb-Threatening Ischemia , Disease Models, Animal , Endothelial Cells/metabolism , Genetic Therapy , Ischemia/genetics , Ischemia/pathology , Ischemia/therapy , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Pathologic/therapy , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Stem Cell Res Ther ; 12(1): 58, 2021 01 13.
Article En | MEDLINE | ID: mdl-33436054

INTRODUCTION: Critical limb ischemia (CLI) is the most advanced form of peripheral arterial disease (PAD) characterized by ischemic rest pain and non-healing ulcers. Currently, the standard therapy for CLI is the surgical reconstruction and endovascular therapy or limb amputation for patients with no treatment options. Neovasculogenesis induced by mesenchymal stem cells (MSCs) therapy is a promising approach to improve CLI. Owing to their angiogenic and immunomodulatory potential, MSCs are perfect candidates for the treatment of CLI. The purpose of this study was to determine and compare the in vitro and in vivo effects of allogeneic bone marrow mesenchymal stem cells (BM-MSCs) and adipose tissue mesenchymal stem cells (AT-MSCs) on CLI treatment. METHODS: For the first step, BM-MSCs and AT-MSCs were isolated and characterized for the characteristic MSC phenotypes. Then, femoral artery ligation and total excision of the femoral artery were performed on C57BL/6 mice to create a CLI model. The cells were evaluated for their in vitro and in vivo biological characteristics for CLI cell therapy. In order to determine these characteristics, the following tests were performed: morphology, flow cytometry, differentiation to osteocyte and adipocyte, wound healing assay, and behavioral tests including Tarlov, Ischemia, Modified ischemia, Function and the grade of limb necrosis scores, donor cell survival assay, and histological analysis. RESULTS: Our cellular and functional tests indicated that during 28 days after cell transplantation, BM-MSCs had a great effect on endothelial cell migration, muscle restructure, functional improvements, and neovascularization in ischemic tissues compared with AT-MSCs and control groups. CONCLUSIONS: Allogeneic BM-MSC transplantation resulted in a more effective recovery from critical limb ischemia compared to AT-MSCs transplantation. In fact, BM-MSC transplantation could be considered as a promising therapy for diseases with insufficient angiogenesis including hindlimb ischemia.


Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Adipose Tissue , Animals , Bone Marrow , Bone Marrow Cells , Cell- and Tissue-Based Therapy , Hindlimb , Humans , Ischemia/therapy , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic
5.
Curr Pharm Des ; 26(38): 4883-4893, 2020.
Article En | MEDLINE | ID: mdl-32364067

BACKGROUND: MicroRNA-126, a microRNA implicated in blood vessel integrity and angiogenesis is significantly up/down regulated in different physiological and pathological conditions related to angiogenesis such as cardiovascular formation and angiogenesis dependent diseases. MicroRNA-126 plays a critical role in angiogenesis via regulating the proliferation, differentiation, migration, and apoptosis of angiogenesis related cells such as endothelial cells. OBJECTIVE: The aim of this review is to investigate the molecular mechanisms and the effects of microRNA-126 on the process of angiogenesis in pathophysiological conditions. METHODS: To conduct this review, related articles published between 2001 and 2019 were collected from the PubMed, Web of Science, Google Scholar, Scopus and Scientific Information Database using search terms such as microRNA-126, angiogenesis, cardiovascular disorders, hypoxia, VEFG-A, endothelial cells, VEGF pathway, and gene silencing. Then, the qualified articles were reviewed. RESULTS: MicroRNA-126 regulates the response of endothelial cells to VEGF, through directly repressing multiple targets, including Sprouty-related EVH1 domain-containing protein 1 (SPRED1) and phosphoinositol-3 kinase regulatory subunit 2 (PIK3R2/p85-b). MicroRNA-126 -3p and microRNA-126 -5p have cell-type and strandspecific functions and also various targets in angiogenesis that lead to the regulation of angiogenesis via different pathways and consequently diverse responses. CONCLUSION: MicroRNA-126 can bind to multiple targets and potentially be both positive and negative regulators of gene expression. Thus, microRNA-126 could cause the opposite biological effects depending on the context. As a result, understanding the different cellular pathways through which microRNA-126 regulates angiogenesis in various situations is a critical aspect in the development of novel and effective treatments for diseases with insufficient angiogenesis.


Endothelial Cells , MicroRNAs , Apoptosis , Cell Proliferation , Humans , MicroRNAs/genetics , Neovascularization, Pathologic/genetics
...