Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Int J Radiat Biol ; 100(2): 209-219, 2024.
Article in English | MEDLINE | ID: mdl-37819928

ABSTRACT

PURPOSE: The study aimed to protect patients' skin against ionizing irradiation during radiotherapy by using astaxanthin-encapsulated nanostructured lipid carriers (NLC-ATX). MATERIALS AND METHODS: NLC-ATX was prepared by a combined method of hot homogenization and sonication. Cytotoxicity of NLC-ATX was evaluated by MTT colorimetric assay. The in vitro radioprotection of NLC-ATX for human fibroblast (HF) cells was investigated based on the level of ROS (reactive oxygen species), DNA damage, and cell death caused by X-irradiation. In addition, the in vivo radioprotection was evaluated based on the appearance and histological structure of the irradiated skin. RESULTS: NLC-ATX was successfully prepared, with a mean particle size, zeta potential, and encapsulation efficiency of 114.4 nm, -34.1 mV, and 85.67%, respectively. Compared to the control, NLC-ATX, at an optimum ATX concentration under in vitro condition, reduced the amount of generated ROS and DNA damage of 81.6% and 41.6%, respectively, after X-radiation, resulting in a significant decrease in cell death by 62.69%. Under in vivo condition, after the 9th day of X-irradiation (equivalent to an accumulated dose of 14 Gy), the dorsal skin of five out of six NLC-ATX-untreated mice exhibited grade-1 skin damage, according to CTCAE v5.0, while treatment with NLC-ATX protected 6/6 mice from acute skin damage. Moreover, on the 28th day after the first X-irradiation, the histological images illustrated that NLC-ATX at an ATX concentration of 0.25 µg/mL exhibited good recovery of the skin, with barely any difference noted in the collagen fibers and sebaceous glands compared to normal skin. CONCLUSIONS: NLC-ATX shows potential for application in skin protection against adverse effects of ionizing rays during radiotherapy.


Subject(s)
Drug Carriers , Skin , Humans , Mice , Animals , Reactive Oxygen Species/metabolism , Drug Carriers/chemistry , Skin/metabolism , Lipids , Particle Size , Xanthophylls
2.
J Microencapsul ; 40(8): 613-629, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37815151

ABSTRACT

This study aims to evaluate the radioprotective effects of liposomes encapsulating curcumin (Lip-CUR), silibinin (Lip-SIL), α-tocopherol (Lip-TOC), quercetin (Lip-QUE) and resveratrol (Lip-RES) in alleviating the adverse effects of ionising irradiation on human lymphoctyes and skin cells in radiotherapy. Liposomes encapsulating the above natural radioprotectants (Lip-NRPs) were prepared by the film hydration method combined with sonication. Their radioprotective effects for the cells against X-irradiation was evaluated using trypan-blue assay and γ-H2AX assay. All prepared Lip-NRPs had a mean diameter less than 240 nm, polydispersity index less than 0.32, and zeta potential more than -23 mV. Among them, the radioprotective effect of Lip-RES was lowest, while that of Lip-QUE was highest. Lip-SIL also exhibited a high radioprotective effect despite its low DPPH-radical scavenging activity (12.9%). The radioprotective effects of Lip-NRPs do not solely depend on the free radical scavenging activity of NRPs but also on their ability to activate cellular mechanisms.


Subject(s)
Curcumin , Liposomes , Humans , Resveratrol , Skin , Curcumin/pharmacology , Lymphocytes
3.
Iran J Basic Med Sci ; 26(4): 429-437, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37009008

ABSTRACT

Objectives: Acute hindlimb ischemia is a peripheral arterial disease that severely affects the patient's health. Injection of stem cells-derived exosomes that promote angiogenesis is a promising therapeutic strategy to increase perfusion and repair ischemic tissues. This study aimed to evaluate the efficacy of adipose stem cell-derived exosomes injection (ADSC-Exos) in treating acute mouse hindlimb ischemia. Materials and Methods: ADSC-Exos were collected via ultracentrifugation. Exosome-specific markers were analyzed via flow cytometry. The morphology of exosomes was detected by TEM. A dose of 100 ug exosomes/100 ul PBS was locally injected into acute mice ischemic hindlimb. The treatment efficacy was evaluated based on the oxygen saturation level, limb function, new blood vessel formation, muscle structure recovery, and limb necrosis grade. Results: ADSC-exosomes expressed high positivity for markers CD9 (76.0%), CD63 (91.2%), and CD81 (99.6%), and have a cup shape. After being injected into the muscle, in the treatment group, many small and short blood vessels formed around the first ligation and grew down toward the second ligation. The SpO2 level, reperfusion, and recovery of the limb function are more positively improved in the treatment group. On day 28, the muscle's histological structure in the treatment group is similar to normal tissue. Approximately 33.33% of the mice had grade I and II lesions and there were no grade III and IV observed in the treatment group. Meanwhile, in the placebo group, 60% had grade I to IV lesions. Conclusion: ADSC-Exos showed the ability to stimulate angiogenesis and significantly reduce the rate of limb necrosis.

4.
Adv Exp Med Biol ; 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-37014542

ABSTRACT

INTRODUCTION: Exosomes derived from mesenchymal stem cells (MSCs) are crucial mediators of the paracrine effects as well as tissue repair and have promising clinical applications. They enhance tissue regeneration by reducing inflammatory responses, enhancing proliferation, inhibiting apoptosis, and stimulating angiogenesis. This study aimed to evaluate the mechanism of angiogenesis supported by exosomes derived from MSCs. METHODS: Exosomes were isolated via ultracentrifugation of a conditioned medium collected from human umbilical cord MSC (hUCMSC) cultures. These exosomes were characterized using transmission electron microscopy, and the expression of specific markers (CD9, CD81, and CD63) was evaluated. To understand the mechanism of angiogenesis, we evaluated the effects of exosomes in endothelial cells (HUVECs). The obtained exosomes were supplemented at a dose of 20 µg/mL into two kinds of culture media for HUVECs (M200 medium and endothelial cell growth medium), while phosphate-buffered saline was added to these media as a control. The effects of the exosomes were evaluated based on the formation of a tubular structure in the culture and the expression of angiogenic genes (MMP-2, Ephrin B2, Ephrin B4, Flk1, Flt1, VWF, VE-cadherin, CD31, ANG1, ANG2, and HGF) via RT-PCR. RESULTS: The exosomes were obtained from the hUCMSCs at a concentration of 0.7 ± 0.029 µg/mL. They accelerated the formation of new blood vessels by upregulating HGF, VWF, CD31, Flt1, and Flk1 (especially VWF and Flt1). CONCLUSION: Exosomes derived from hUCMSCs can promote angiogenesis through upregulation of VWF and Flt1 in endothelial cells.

5.
Adv Exp Med Biol ; 2023 Mar 30.
Article in English | MEDLINE | ID: mdl-36991295

ABSTRACT

INTRODUCTION: Recent studies have demonstrated that adipose tissue-derived stem cell (ADSC) transplantation could promote neoangiogenesis in various ischemic diseases. However, as whole cells, ADSCs have some disadvantages, such as shipping and storage issues, high costs, and controversies related to the fates of grafted cells in the recipients. Therefore, this study aimed to investigate the effects of intravenously infused exosomes purified from human ADSCs on ischemic disease in a murine hindlimb ischemia model. METHODS: ADSCs were cultured in exosome-free medium for 48 h before the conditioned medium was collected for exosome isolation by ultracentrifugation. The murine ischemic hindlimb models were created by cutting and burning the hindlimb arteries. Exosomes were intravenously infused into murine models (ADSC-Exo group), with phosphate-buffered saline (PBS) used as a placebo (PBS group). Treatment efficacy was determined using a murine mobility assay (frequency of pedaling in water per 10 s), peripheral blood oxygen saturation (SpO2 index), and the recovery of vascular circulation by trypan blue staining. The formation of blood vessels was shown by X-ray. Expression levels of genes related to angiogenesis and muscle tissue repair were quantified by quantitative reverse-transcription polymerase chain reaction. Finally, H&E staining was used to determine the histological structure of muscle in the treatment and placebo groups. RESULTS: The rates of acute limb ischemia in the PBS and ADSC-Exo injection groups were 66% (9/16 mice) and 43% (6/14 mice), respectively. The mobility of the limbs 28 days after surgery was significantly different between the ADSC-Exo treatment group (41 ± 1 times/10 s) and the PBS group (24 ± 1 times/10 s; n = 3; p < 0.05). Peripheral blood oxygen saturation 21 days after treatment was 83.83% ± 2.02% in the PBS group and 83% ± 1.73% in the ADSC-Exo treatment group, and the difference was not statistically significant (n = 3, p > 0.05). On day 7 after treatment, the time required to stain the toes after trypan blue injection was 20.67 ± 12.5 s and 85 ± 7.09 s in the ADSC-Exo and PBS groups, respectively (n = 3, p < 0.05). On day 3 after the operation, the expression of genes promoting angiogenesis and muscle remodeling, such as Flk1, Vwf, Ang1, Tgfb1, Myod, and Myf5, was increased 4-8 times in the ADSC-Exo group compared with the PBS group. No mice in either group died during the experimental period. CONCLUSIONS: These results revealed that intravenous infusion of human ADSC-derived exosomes is a safe and effective method to treat ischemic disease, especially hindlimb ischemia, by promoting angiogenesis and muscle regeneration.

6.
Adv Exp Med Biol ; 2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35389201

ABSTRACT

INTRODUCTION: In recent years, both stromal vascular fraction (SVF) from adipose tissue and mesenchymal stem cells (MSC) from adipose tissues were extensively used in both preclinical and clinical treatment for various diseases. Some studies reported differences in treatment efficacy between SVFs and MSCs in animals as well as in humans. Therefore, this study is aimed to evaluate the immune modulation and angiogenic potential of SVFs and MSCs from the same SVF samples to support an explanation when SVFs or MSCs should be used. METHODS: The adipose tissue samples from ten female donors with consent forms were collected. SVFs from these samples were isolated according to the published protocols. The existence of mesenchymal cells that positive with CD44, CD73, CD90, and CD105 and endothelial progenitor cells that positive with CD31 and CD34 was determined using flow cytometry. Three samples of SVFs with similar percentages of mesenchymal cell portion and endothelial progenitor cell portion were used to isolate MSCs. Obtained MSCs were confirmed as MSCs using the ISCT minimal criteria. To compare the immune modulation of SVF and MSCs, the mixed lymphocyte assay was used. The lymphocyte proliferation, as well as IFN-gamma and TNF-alpha concentrations, were determined. To compare the angiogenic potential, the angiogenesis in quail embryo assay was used. The angiogenesis efficacy was measured based on the vessel areas formed in the embryos after 7 days. RESULTS: The results showed that all SVF samples contained the portions of mesenchymal cells and endothelial progenitor cells. MSCs from SVFs meet all minimal criteria of MSCs that suggested by ISCT. MSCs from SVFs efficiently suppressed the immune cell proliferation compared to the SVFs, especially at ratios of 1:4 (1 MSCs: 4 immune cells). MSCs also inhibited the IFN-gamma and TNF-alpha production more efficiently than SVFs (p < 0.05). However, in quail embryo models, SVFs triggered the angiogenesis and neovessel formation better than MSCs with more significant vessel areas after 7 days (p < 0.05). CONCLUSION: This study suggested that SVFs and MSCs have different potentials for immune modulation and angiogenesis. SVFs help the angiogenesis better than MSCs, while MSCs displayed the more significant immune modulation. These results can guide the usage of SVFs or MSCs in disease treatment.

7.
Adv Exp Med Biol ; 2021 Nov 06.
Article in English | MEDLINE | ID: mdl-34739719

ABSTRACT

INTRODUCTION: The damaged articular cartilage has limited self-regeneration capacity because of the absence of blood vessels, lymphatics, and nerves. Cartilage transplantation is, hence, a popular method used to treat this disease. However, sources of autograft and allogenic cartilage for transplantation are limited. Therefore, this study aims to suggest a simple method to produce engineered cartilage from human adipose-derived mesenchymal stem cells (ADSCs) and poly (ε-caprolactone) (PCL) scaffolds. METHODS: ADSCs were isolated and expanded from fat tissues according to published protocols. PCL-porous scaffolds were produced from PCL with 5 × 5 × 0.6 mm3 with 200-400 µ m pore sizes. ADSCs were seeded on the PCL scaffolds at three different densities (104, 105, 106 cells per scaffold). The adherence of ADSCs on the surface of PCL scaffolds was evaluated based on an immunostaining assay to determine the presence of ADSCs. The cell proliferation on PCL scaffolds was determined by MTT assay. The complexity in ADSCs and PCL scaffolds was induced to cartilage using a chondrogenesis medium. The engineered cartilage was characterized by the accumulation of proteoglycan and aggrecan by Safranin O staining assay. Their structures were evaluated using an H-E staining assay. Finally, these engineered cartilage tissues were transplanted into mice to assess cartilage maturation when compared to natural cartilage. RESULTS: The results showed that the engineered cartilage tissues could be successfully produced by cultures of ADSCs on poly ε-caprolactone scaffolds in combination with chondrogenesis medium. The suitable density of ADSCs was 106 cells/per scaffold of 5 × 6 × 0.6 mm3 with pore size from 200 to 400 µ m. CONCLUSION: The results showed that an in vitro cartilage tissue was created from ADSCs and PCL scaffold. The cartilage tissue exists in the mice for 6 months.

8.
Adv Exp Med Biol ; 2021 Nov 06.
Article in English | MEDLINE | ID: mdl-34739721

ABSTRACT

INTRODUCTION: After injuries, the cartilage healing capacity is limited owing to its nature as a particular connective tissue without blood vessels, lymphatics, or nerves. The creation of artificial cartilage tissue mimics the biological properties of native cartilage and can reduce the need for donated tissue. Fibrin is a type of biodegradable scaffold that has great potential in tissue engineering applications. It can become good material for cell adhesion and proliferation in vitro. Therefore, this study aimed to create a cartilage tissue in vitro using umbilical cord-derived mesenchymal stem cells (UCMSC) and growth factor-rich fibrin (GRF) scaffolds. METHODS: UCMSCs were isolated and expanded, and platelet-rich plasma (PRP) preparations were performed following previously published protocols. PRP was activated (aPRP) by a 0.45-µm syringe filter to release growth factors inside the platelets. Each 2.105 of the UCMSCs were suspended in 2 ml of aPRP to make the mixture of MSC and PRP (MSC-PRP). Then, Ca2+ solution was added to this mixture to produce the fibril scaffold with UCMSCs inside. UCMSCs' adhesion and proliferation inside the scaffold were evaluated by observation under inverted microscopy, H-E staining, MTT assays, and scanning electron microscopy (SEM). The fibril structure containing UCMSCs was cultured, and chondrogenesis was induced using commercial chondrogenesis media for 21 days (iMSC-GRF). The differentiation in efficacy toward cartilage was evaluated based on the accumulation of aggrecan (acan), glycosaminoglycans (GAGs), and collagen type II (Col II). RESULTS: The results showed that we successfully created a cartilage tissue with some characteristics that mimic the properties of natural cartilage. The engineered cartilage tissue was positive with some cartilage protein, such as acan, GAG, and Coll II. In vitro cartilage presented some natural chondrocyte-like cells. The artificial cartilage tissue was positive for CD14, CD34, CD90, CD105, and HLA-DR and negative for CD44, CD45, and CD73. CONCLUSION: These results showed that using UCMSCs and growth factor-rich fibril from platelet-rich plasma was feasible to produce engineered cartilage tissue for further experiments or clinical usage.

9.
Adv Exp Med Biol ; 2021 Nov 06.
Article in English | MEDLINE | ID: mdl-34739722

ABSTRACT

INTRODUCTION: Articular cartilage is limited in self-repair following injuries due to avascular, lymphatic, and nerve absence. Recent treatments for cartilage injuries, such as physical therapy, anti-inflammatory medication, chondrocyte implantation, and joint replacement, still have limitations. This study aimed to evaluate the treatment efficacy of human umbilical cord-derived mesenchymal stem cell sheet (UCMSCS) transplantation in rat models of the osteochondral femoral head defect. METHODS: Models of osteochondral femoral head defect were produced in rats by drilling in order to reach the femoral bone tissue through the cartilage layer. Then, UCMSCS was implanted in the created cartilage lesion. The treatment efficacy was monitored by X-ray imaging. The cartilage regeneration was evaluated based on the hematoxylin and eosin staining, and proteoglycan accumulation was detected by staining Safranin O and Fast Green. The physiological, weight, or movement activity of rats were recorded during the treatment period. RESULTS: UCMSCS transplantation showed positive effects on the cartilage regeneration in osteochondral femoral head defect grade 4 (according to ICRS score/grade). Particularly, after 12 weeks of implantation of UCMSCS, the defect was filled with hyaline cartilage-like cells and accumulated a large density of proteoglycan. The osteochondral defect score significantly increased in the treated rats compared to the untreated rats (11.67 ± 0.6 and 9.67 ± 0.6, respectively) (p < 0.05). The histological score also increased in treated rats compared to untreated rats (21.33 ± 1.53 vs. 18.00 ± 1.00) (p < 0.0001). The accumulation of proteoglycan was higher in treated rats (20.50 ± 2.23) than untreated rats (5.38 ± 0.36) (p < 0.05). There was no change in the physiological activities between treated and untreated rats recorded during the study. CONCLUSION: MSCS transplantation could promote regeneration in advanced cartilage injury.

10.
Tissue Eng Regen Med ; 18(4): 587-611, 2021 08.
Article in English | MEDLINE | ID: mdl-33884577

ABSTRACT

Ischemic diseases are conditions associated with the restriction or blockage of blood supply to specific tissues. These conditions can cause moderate to severe complications in patients, and can lead to permanent disabilities. Since they are blood vessel-related diseases, ischemic diseases are usually treated with endothelial cells or endothelial progenitor cells that can regenerate new blood vessels. However, in recent years, mesenchymal stem cells (MSCs) have shown potent bioeffects on angiogenesis, thus playing a role in blood regeneration. Indeed, MSCs can trigger angiogenesis at ischemic sites by several mechanisms related to their trans-differentiation potential. These mechanisms include inhibition of apoptosis, stimulation of angiogenesis via angiogenic growth factors, and regulation of immune responses, as well as regulation of scarring to suppress blood vessel regeneration when needed. However, preclinical and clinical trials of MSC transplantation in ischemic diseases have shown some limitations in terms of treatment efficacy. Such studies have emphasized the current challenges of MSC-based therapies. Treatment efficacy could be enhanced if the limitations were better understood and potentially resolved. This review will summarize some of the strategies by which MSCs have been utilized for ischemic disease treatment, and will highlight some challenges of those applications as well as suggesting some strategies to improve treatment efficacy.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Endothelial Cells , Humans , Ischemia/therapy , Regeneration
11.
World J Stem Cells ; 12(8): 721-730, 2020 Aug 26.
Article in English | MEDLINE | ID: mdl-32952854

ABSTRACT

Coronavirus disease-2019 (COVID-19) has affected more than 200 countries worldwide. This disease has hugely affected healthcare systems as well as the economy to an extent never seen before. To date, COVID-19 infection has led to about 165000 deaths in 150 countries. At present, there is no specific drug or efficient treatment for this disease. In this analysis based on evidential relationships of the biological characteristics of MSCs, especially umbilical cord (UC)-derived MSCs as well as the first clinical trial using MSCs for COVID-19 treatment, we discuss the use of UC-MSCs to improve the symptoms of COVID-19 in patients.

12.
Adv Exp Med Biol ; 1084: 207-220, 2019.
Article in English | MEDLINE | ID: mdl-31214911

ABSTRACT

INTRODUCTION: Human umbilical cord-derived mesenchymal stem cells (UCMSCs) are multiple potential stem cells that can differentiate into various kinds of functional cells, including adipocytes, osteoblasts, and chondroblasts. Thus, UCMSCs have recently been used in both stem cell therapy and tissue engineering applications to produce various functional tissues. This study aimed to evaluate the proliferation and differentiation of UCMSCs on porous scaffolds. METHODS: UCMSCs were established in a previous study and kept in liquid nitrogen. They were thawed and expanded in vitro to yield enough cells for further experiments. The cells were characterized as having MSC phenotype. They were seeded onto culture medium-treated porous scaffolds or on non-treated porous scaffolds at different densities of UCMSCs (105, 2.1 × 105, and 5 × 105 cells/0.005 g scaffold). The existence of UCMSCs on the scaffold was evaluated by nucleic staining using Hoechst 33342 dye, while cell proliferation on the scaffold was determined by MTT assay. Osteogenic differentiation was evaluated by changes in cellular morphology, accumulation of extracellular calcium, and expression of osteoblast-specific genes (including runx2, osteopontin (OPN), and osteocalcin (OCN)). RESULTS: The data showed that UCMSCs could attach, proliferate, and differentiate on both treated and non-treated scaffolds but were better on the treated scaffold. At a cell density of 105 cells/0.005 g scaffold, the adherent and proliferative abilities of UCMSCs were higher than that of the other densities after 14 days of culture (p < 0.05). Adherent UCMSCs on the scaffold could be induced into osteoblasts in the osteogenic medium after 21 days of induction. These cells accumulated calcium in the extracellular matrix that was positive with Alizarin Red staining. They also expressed some genes related to osteoblasts, including runx2, OPN, and OCN. CONCLUSION: UCMSCs could adhere, proliferate, and differentiate into osteoblasts on porous scaffolds. Therefore, porous scaffolds (such as Variotis) may be suitable scaffolds for producing bone tissue in combination with UCMSCs.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Tissue Scaffolds , Cell Proliferation , Cells, Cultured , Humans , Tissue Engineering/standards , Tissue Scaffolds/standards , Umbilical Cord/cytology
13.
J Microencapsul ; 36(2): 156-168, 2019 Mar.
Article in English | MEDLINE | ID: mdl-31030591

ABSTRACT

This study aimed to compare the in vivo effectiveness between curcumin-oligochitosan nanoplexes (CUR-OCH nanoplexes) and oligochitosan-coated curcumin-encapsulated liposomes (OCH-Lip-CUR) with respect to wound healing and scar treatment. Firstly, CUR-OCH nanoplexes was prepared by drug-polysaccharide complexation method and OCH-Lip-CUR was prepared by a combining method of lipid-film hydration and sonication. Their in vitro cytotoxicity and in vivo wound healing and scar treatment effectiveness were evaluated using 3T3 cells and mice Mus musculus var. Albino, respectively. The resutls indicated that both of them were in nanosize with a moderate PDI (less than 0.3), and exhibited negligible cytotoxicity at low CUR concentration (0.01 mg/mL). Moreover, their application onto wounds resulted in faster healing and higher scar treatment effectiveness than control samples. Interestingly, OCH-Lip-CUR exhibited higher in vivo effectiveness than CUR-OCH nanoplexes. However, based on their own advantages, both of them were good candidates for a commercial formulation for wound healing and scar treatment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cicatrix/drug therapy , Curcumin/administration & dosage , Wound Healing/drug effects , 3T3 Cells , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chitin/analogs & derivatives , Chitin/chemistry , Chitosan , Curcumin/analogs & derivatives , Curcumin/therapeutic use , Drug Liberation , Liposomes/chemistry , Male , Mice , Nanoconjugates/chemistry , Oligosaccharides
14.
Mater Sci Eng C Mater Biol Appl ; 98: 54-64, 2019 May.
Article in English | MEDLINE | ID: mdl-30813056

ABSTRACT

While the wound healing activity of curcumin (CUR) has been well-established, its clinical effectiveness remains limited due to the inherently low aqueous CUR solubility, resulting in suboptimal CUR exposure in the wound sites. Previously, we developed high-payload amorphous nanoparticle complex (or nanoplex) of CUR and chitosan (CHI) capable of CUR solubility enhancement by drug-polyelectrolyte complexation. The CUR-CHI nanoplex, however, exhibited poor colloidal stability due to its strong agglomeration tendency. Herein we hypothesized that the colloidal stability could be improved by replacing CHI with its oligomers (OCHI) owed to the better charge distribution in OCHI. The effects of key parameters in drug-polyelectrolyte complexation (i.e. pH, salt inclusion, CUR concentration, and OCHI/CUR charge ratio) on the physical characteristics and preparation efficiency of the CUR-OCHI nanoplex produced were investigated. The in vivo wound healing efficacy of the CUR-OCHI nanoplex and its cytotoxicity towards human keratinocytes cells were examined. The results showed that CUR-OCHI nanoplex exhibited prolonged colloidal stability (72 h versus <24 h for the CUR-CHI nanoplex). At the optimal condition, the CUR-OCHI nanoplex (without ultrasonication) exhibited size, zeta potential, and CUR payload of ≈140 nm, 20 mV, and 78% (w/w), respectively. The nanoplex preparation was simple yet robust at nearly 100% CUR utilization rate. The CUR-OCHI nanoplex exhibited superior wound healing efficacy to the native CUR with wound closure of >90% after 7 days versus 9 days for the native CUR resulting in smaller scars, attributed to its generation of high CUR concentration in the wound sites.


Subject(s)
Chitin/analogs & derivatives , Chitosan/chemistry , Curcumin/chemistry , Curcumin/pharmacology , Nanoparticles/chemistry , Cells, Cultured , Chitin/chemistry , Drug Carriers/chemistry , Humans , Keratinocytes/drug effects , Oligosaccharides , Wound Healing/drug effects
15.
Adv Exp Med Biol ; 1084: 45-60, 2019.
Article in English | MEDLINE | ID: mdl-29299874

ABSTRACT

In vitro production of tissues or tissue engineering is a promising approach to produce artificial tissues for regenerative medicine. There are at least three important components of tissue engineering, including stem cells, scaffolds and growth factors. This study aimed to produce cartilage tissues in vitro from culture and chondrogenic differentiation of rabbit bone marrow-derived mesenchymal stem cells (BMMSCs), induced by chondrogenesis medium, on biodegradable polycaprolactone (PCL) scaffolds. BMMSCs were isolated from rabbit bone marrow according to the standard protocol. The adherence, proliferation and differentiation of BMMSCs on scaffolds were investigated using two scaffold systems: PCL scaffolds and collagen-coated PCL (PCL/col) scaffolds. The results showed that BMMSCs could attach and grow on both PCL and PCL/col scaffolds. However, the adhesion efficacy of BMMSCs on the PCL/col scaffolds was significantly better than on PCL scaffolds. Under induced conditions, BMMSCs on PLC/col scaffolds showed increased aggrecan accumulation and upregulated expression of chondrogenesis-associated genes (e.g. collagen type II, collagen type I, aggrecan and collagen type X) after 3, 7, 21 and 28 days of induction. These in vitro cartilage tissues could form mature chondrocyte-like cells after they were grafted into rabbits. The results suggest that use of BMMSCs in combination with polycaprolactone scaffolds and chondrogenesis medium can be a way to form in vitro cartilage tissue.


Subject(s)
Bone Marrow , Chondrogenesis , Mesenchymal Stem Cells , Polyesters , Tissue Scaffolds , Animals , Cartilage/cytology , Cells, Cultured , Mesenchymal Stem Cells/cytology , Polyesters/chemistry , Rabbits , Tissue Engineering , Tissue Scaffolds/chemistry
16.
Adv Exp Med Biol ; 1083: 1-17, 2018.
Article in English | MEDLINE | ID: mdl-28687961

ABSTRACT

Cell migration and molecular mechanisms during healing of damaged vascular or muscle tissues are emerging fields of interest worldwide. The study herein focuses on evaluating the role of allogenic adipose-derived mesenchymal stem cells (ADMSCs) in restoring damaged tissues. Using a hindlimb ischemic mouse model, ADMSC-mediated induction of cell migration and gene expression related to myocyte regeneration and angiogenesis were evaluated. ADMSCs were labeled with GFP (ADMSC-GFP). The proximal end of the femoral blood vessel of mice (over 6 months of age) are ligated at two positions then cut between the two ties. Hindlimb ischemic mice were randomly divided into two groups: Group I (n = 30) which was injected with PBS (100 µL) and Group II (n = 30) which was transplanted with ADMSC-GFP (106 cells/100 µL PBS) at the rectus femoris muscle. The migration of ADMSC-GFP in hindlimb was analyzed by UV-Vis system. The expression of genes related to angiogenesis and muscle tissue repair was quantified by real-time RT-PCR. The results showed that ADMSCs existed in the grafted hindlimb for 7 days. Grafted cells migrated to other damaged areas such as thigh and heel. In both groups the ischemic hindlimb showed an increased expression of several angiogenic genes, including Flt-1, Flk-1, and Ang-2. In particular, the expression of Ang-2 and myogenic-related gene MyoD was significantly increased in the ADMSC-treated group compared to the PBS-treated (control) group; the expression increased at day 28 compared to day 3. The other factors, such as VE-Cadherin, HGF, CD31, Myf5, and TGF-ß, were also more highly expressed in the ADMSC-treated group than in the control group. Thus, grafted ADMSCs were able to migrate to other areas in the injured hindlimb, persist for approximately 7 days, and have a significantly positive impact on stimulating expression of myogenic- and angiogenesis-related genes.


Subject(s)
Ischemia/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Adipose Tissue/cytology , Animals , Hindlimb , Mice , Random Allocation
17.
Cytotechnology ; 69(5): 801-814, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28466428

ABSTRACT

Ischemia are common conditions related to lack of blood supply to tissues. Depending on the ischemic sites, ischemia can cause different diseases, such as hindlimb ischemia, heart infarction and stroke. This study aims to evaluate how extracellular vesicles (EVs) derived from ETV2 transfected fibroblasts affect endothelial cell proliferation and neovascularization in a murine model of hindlimb ischemia. Human fibroblasts were isolated and cultured under standard conditions and expanded to the 3th passage before use in experiments. Human fibroblasts were transduced with a viral vector containing the ETV2 gene. Transduced cells were selected by puromycin treatment. These cells were further cultured for collection of EVs, which were isolated from culture supernatant. Following co-culture with endothelial cells, EVs were evaluated for their effect on endothelial cell proliferation and were directly injected into ischemic tissues of a murine model of hindlimb ischemia. The results showed that EVs could induce endothelial cell proliferation in vitro and improved neovascularization in a murine model of hindlimb ischemia. Our results suggest that EVs derived from ETV2-transfected fibroblasts can be promising non-cellular products for the regeneration of blood vessels.

18.
In Vitro Cell Dev Biol Anim ; 53(7): 616-625, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28424975

ABSTRACT

Ischemia is the reduction of blood flow to tissues by injury of blood vessels. Depending on the sites of tissues and grade of ischemia, ischemia can cause many serious complications. This study aimed to evaluate the effects of the E-twenty six (ETS) factor Ets variant 2 (ETV2) gene expression in angiogenesis and the effect of ETV2 gene therapy in a mouse model of hindlimb ischemia. The role of ETV2 on endothelial cell proliferation was evaluated in vitro. Knockdown of ETV2 expression was done using short hairpin RNA (shRNA) lentiviral viral particles. The ETV2 viral vector was injected into the skeletal muscles at the ligated and burned sites of the hindlimb and evaluated for its efficacy as a gene therapy modality for ischemia. Vascular regeneration in mice was indirectly evaluated by changes in mouse survival, necrotic grades of the leg, normal blood oxygen saturation level (SpO2), and blood flow by trypan blue injection assay. Preliminary data showed that ETV2 expression played a role in angiogenesis of endothelial cells. ETV2 overexpression could trigger and stimulate proliferation of skeletal endothelial cells. In vivo knockdown of ETV2 expression inhibited the auto-recovery of ischemic hindlimb, while overexpression of ETV2 helped to rescue leg loss and reduce necrosis, significantly improving angiogenesis in hindlimb ischemia. Our findings demonstrate that ETV2 gene therapy is a potentially effective modality for vascular regeneration.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/pathology , Hindlimb/blood supply , Hindlimb/pathology , Ischemia/pathology , Transcription Factors/metabolism , Acute Disease , Animals , Cell Hypoxia , Cell Proliferation , Cell Separation , Cells, Cultured , Disease Models, Animal , Ischemia/metabolism , Lentivirus/metabolism , Mice , Muscles/pathology , Necrosis
19.
Stem Cells Transl Med ; 6(1): 187-195, 2017 01.
Article in English | MEDLINE | ID: mdl-28170179

ABSTRACT

Osteoarthritis (OA) is a degenerative cartilage disease that is characterized by a local inflammatory reaction. Consequently, many studies have been performed to identify suitable prevention and treatment interventions. In recent years, both arthroscopic microfracture (AM) and stem cell therapy have been used clinically to treat OA. This study aimed to evaluate the clinical effects of AM in the presence and absence of a stromal vascular fraction (SVF) injection in the management of patients with OA. Thirty patients with grade 2 or 3 (Lawrence scale) OA of the knee participated in this study. Placebo group patients (n = 15) received AM alone; treatment group patients (n = 15) received AM and an adipose tissue-derived SVF injection. The SVF was suspended in platelet-rich plasma (PRP) before injection into the joint. Patient groups were monitored and scored with the Western Ontario and McMaster Universities Arthritis Index (WOMAC), Lysholm, Visual Analog Pain Scale (VAS), and modified Outerbridge classifications before treatment and at 6, 12, and 18 months post-treatment. Bone marrow edema was also assessed at these time points. Patients were evaluated for knee activity (joint motion amplitude) and adverse effects relating to surgery and stem cell injection. Treatment efficacy was significantly different between placebo and treatment groups. All treatment group patients had significantly reduced pain and WOMAC scores, and increased Lysholm and VAS scores compared with the placebo group. These findings suggest that the SVF/PRP injection efficiently improved OA for 18 months after treatment. This study will be continuously monitored for additional 24 months. Stem Cells Translational Medicine 2017;6:187-195.


Subject(s)
Arthroscopy , Fractures, Stress/pathology , Osteoarthritis, Knee/therapy , Bone Marrow/pathology , Edema/pathology , Female , Fractures, Stress/physiopathology , Humans , Injections , Knee Joint/pathology , Knee Joint/physiopathology , Magnetic Resonance Imaging , Male , Middle Aged , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/physiopathology , Stromal Cells/metabolism , Treatment Outcome , Visual Analog Scale
20.
In Vitro Cell Dev Biol Anim ; 53(3): 207-216, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27778229

ABSTRACT

Endothelial progenitor cells (EPCs) play an important role in angiogenesis. However, they exist in limited numbers in the human body. This study was aimed to produce EPCs, for autologous transplantation, using direct reprogramming of skin fibroblasts under GMP-compliant conditions. Fibroblasts were collected and cultured from the skin in DMEM/F12 medium supplemented with 5% activated platelet-rich plasma and 1% antibiotic-antimycotic solution. They were then transfected with mRNA ETV2 and incubated in culture medium under hypoxia (5% oxygen) for 14 d. Phenotype analysis of transfected cells confirmed that single-factor ETV2 transfection successfully reprogrammed dermal fibroblasts into functional EPCs. Our results showed that ETV2 mRNA combined with hypoxia can give rise to functional EPCs. The cells exhibited functional phenotypes similar to endothelial cells derived from umbilical cord vein; they expressed CD31 and VEGFR2, and formed capillary-like structures in vitro. Moreover, these EPCs could significantly improve hindlimb ischemia in mouse models. Although the direct conversion efficacy was low (3.12 ± 0.98%), altogether our study demonstrates that functional EPCs can be produced from fibroblasts and can be used in clinical applications.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Endothelial Progenitor Cells/cytology , Neovascularization, Physiologic/genetics , Animals , Cell Hypoxia , Cell Proliferation/genetics , Fibroblasts/cytology , Hindlimb/cytology , Hindlimb/growth & development , Humans , Mice , Skin/cytology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...