Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Adipocyte ; 12(1): 2237164, 2023 12.
Article in English | MEDLINE | ID: mdl-37488770

ABSTRACT

Brown adipocytes were proposed to reverse metabolic conditions such as obesity and diabetes, which make them potential for therapeutic applications. Brown adipocytes and browning process are capable of thermogenesis, the uncoupling metabolism which allows them to promote balanced energy expenditure, a fundamental mechanism for improving metabolic disorders. Thermogenesis process is not only performed by the thermogenin UCPs within the mitochondria, but instead, is globally regulated within brown and browning adipose tissues, which induces signalling molecules that can be sent to nearby and distant tissues to generate systemic effects on metabolism. This review highlights thermogenesis and describes the crosstalk between different organelles within browning and brown adipocytes, as well as their interorgan axes to regulate whole body metabolism. Finally, browning and thermogenesis activation will also be discussed in terms of physiological conditions, in which, we propose that thermogenesis and functional activities of brown adipocytes should be considered individually in future clinical application.


Subject(s)
Adipocytes, Brown , Mitochondria , Energy Metabolism , Adipose Tissue , Thermogenesis
2.
Adv Exp Med Biol ; 2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37291444

ABSTRACT

INTRODUCTION: The pro-inflammatory cytokine interferon-gamma (IFN-γ) is reported to be an agent that boosts the immune modulation of mesenchymal stem cells (MSCs). However, the effects of IFN-γ on the chondrogenic potential of treated MSCs have not been evaluated in depth. This study aimed to evaluate the effects of IFN-γ on the immune modulation and chondrogenic potential of human umbilical cord-derived MSCs (hUC-MSCs). METHODS: UC-MSCs were isolated and expanded following published protocols. They were characterized as MSCs before their use in further experiments. The UC-MSCs were treated with IFN-γ at 10 ng/mL for 48 h. Changes in phenotype were investigated based on changes in MSC markers, immunomodulatory genes (TGF-ß, IL-4, and IDO) for immune modulation, and cartilage-related genes during the induction of differentiation (Col1a2, Col2a1, Sox9, Runx2, and Acan) for chondrogenic potential. RESULTS: IFN-γ-treated UC-MSCs maintained MSC markers and exhibited decreased expression of transcriptional regulatory factors in chondrogenesis (Sox9 and Runx2) and the extracellular matrix-specific genes Col1a2 and Acan but not Col2a1 compared to non-treated cells (p < 0.05). Furthermore, the immunomodulatory capability of IFN-γ-treated UC-MSCs was clearly revealed through their increased expression of IDO and IL-4 and decreased expression of TGF-ß compared to non-treated cells (p < 0.05). CONCLUSION: This study demonstrated that UC-MSCs treated with IFN-γ at 10 ng/mL had reduced expression of chondrocyte-specific genes; however, they maintained multi-lineage differentiation and exhibited immunomodulatory properties.

3.
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.

4.
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.

5.
Cell Tissue Bank ; 24(1): 139-152, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35792988

ABSTRACT

Immune cell therapy has been incorporated into cancer therapy over the past few years. Chimeric antigen receptor T cells (Car-T cells) transplantation is a novel and promising therapy for cancer treatment and introduces a new age of immune cell therapy. However, the expensive nature of genetic modification procedures limits the accessibility of Car-T cells for cancer treatment. Cytokine-induced killer cells (CIKs) can kill the target cells in an MHC-non-restricted manner; these cells can be developed to "off-the-shelf" immune cell products for cancer treatment. However, the anti-tumor potency of freshly thawed CIKs is not well documented. This study aimed to fill this gap, evaluating the anti-tumor potency of freshly thawed CIKs compared to that of freshly cultured CIKs. CIKs were produced from the human umbilical cord blood in accordance with published protocols. CIKs were cryopreserved in xeno-free cryomedium that contains 5% DMSO, 10% human serum in phosphate buffer saline at - 86 °C. These cells were thawed and immediately utilized in assays (called freshly thawed CIKs) with freshly cultured cells are control. The expression of the surface markers of CIKs, cytokine production, and in vitro anti-tumor cytotoxic cells of freshly thawed CIKs were evaluated and compared to freshly cultured CIKs. Additionally, the freshly thawed CIKs were injected into the breast of tumor-bearing mice to assess the anti-tumor potency in vivo. The results obtained in freshly thawed CIKs and freshly cultured CIKs demonstrated that the expression of CD3, and CD56 were comparable in both cases. The production of TNF-α, IFN-γ, and IL-10 was slightly reduced in freshly thawed cells compared to the freshly cultured cells. The in vitro lysis toward MCF-7 cancer cells was similar between freshly thawed and freshly cultured CIKs. Moreover, the freshly thawed CIKs displayed anti-breast tumor activity in the breast tumor-bearing mice. The volume of tumors significantly reduced in the mice grafted with freshly thawed CIKs while, conversely, the tumor volume in mice of the placebo group gradually increased. This study substantiated that freshly thawed CIKs preserved their anti-tumor potency in both in vitro and in vivo conditions. The results initially revealed the great potential of UCB-CIKs for "off-the-shelf" CIK product manufacturing. However, further studies on the effects of cryomedia, freezing rate, and thawing procedure should be undertaken before freshly thawed off-the-shelf UCB-CIKs are utilized in clinical trials.


Subject(s)
Cytokine-Induced Killer Cells , Neoplasms , Animals , Humans , Mice , Cell Proliferation , Cells, Cultured , Fetal Blood , Neoplasms/pathology
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-34739720

ABSTRACT

INTRODUCTION: Adipose-derived stem cells (ADSCs) are mesenchymal stem cells (MSCs) that are found in adipose tissues, which are easily obtained from liposuction procedures using an enzyme mixture. The adhering cells are then selectively cultivated. ADSCs have great potential in regenerative medicine because they are plentiful, easily accessible, and less invasive. They also have an impressive proliferation ability and can be differentiated into mesenchymal lineages and trans-differentiating into many other cell types. In particular, they have extraordinary abilities in immunomodulation. This study aimed to investigate the effects of culture conditions (hypoxia, starvation, and TNF-α treatment) on the immunomodulation of human ADSCs. METHODS: Human ADSCs were expanded in vitro in the standard condition before they were cultured in different stress conditions. ADSCs from passages fifth were confirmed as MSCs by some standard assays suggested by the International Society for Cell and Gene Therapy. These MSCs were used to culture in four different stress conditions: hypoxia, serum starvation, and TNF-α treatment in 48 h. After treatments, MSCs were used to evaluate their immunomodulation capacity using MSCs mixed lymphocyte reaction assay, and the concentrations of IDO, PGE2, IL-6, and IL-10 were secreted in the culture medium. RESULTS: In different stress conditions, ADSCs exhibited different responses related to their immunomodulation. In serum starvation, ADSCs exerted a strong secretion of IDO and PGE2, whereas they showed strong IL-6 secretion in the TNF-α-supplemented medium. When exposed to lymphocytes, ADSCs caused an increase in the ratio of regulatory T cells (Tregs), and co-culture lymphocytes with ADSCs induced in hypoxic malnutrition conditions increased the IL-10 level the most. In addition, when exposed to dendritic cells (DCs), ADSCs inhibited the mature marker expressions of the DCs. CONCLUSION: The current research showed that ADSCs change their immunomodulation properties to survive in in vitro culture environments. Treatment of ADSCs in the starvation medium for 48 h can increase the immunomodulation of ADSCs.

8.
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.

9.
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
10.
J Clin Med ; 10(2)2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33467167

ABSTRACT

Ocular chemical and thermal burns are frequent causes of hospitalization and require immediate interventions and care. Various surgical and pharmacological treatment strategies are employed according to damage severity. Controlling inflammation and neovascularization while promoting normal ocular surface anatomy and function restoration is the principal aim. In the most severe cases, when epithelial healing is severely affected, reconstruction of the ocular surface may be a valid option, which, however, requires expertise, adequate instruments, and qualified donors. Numerous endogenous and exogenous strategies have been considered for corneal repair. Among these, stem cells and their derivatives have offered numerous attractive possibilities in finding an effective way in stimulating corneal regeneration. Limbal epithelial stem cells and mesenchymal cells from the ocular tissue as well as from various sources have demonstrated their effectiveness in dampening neovascularization, scarring, and inflammation, while promoting epithelialization of the injured cornea. Moreover, a plethora of cytokines and growth factors, and extracellular vesicles, which constitute the secretome of these cells, work in concert to enhance wound healing. In this review, we provide an update on the recent potential therapeutic avenues and clinical applications of stem cells and their products in corneal regeneration after burn injury, as well as current imaging strategies for monitoring therapeutic efficacy and damage resolution.

11.
Front Biosci (Landmark Ed) ; 26(2): 266-285, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33049670

ABSTRACT

This study suggested a new method to produce the in vitro cartilage tissues by cartilage differentiation in spheroids from adipose-derived stem cells on porous scaffolds. Adipose-derived stem cells (ADSCs) were used to produce spheroids by the hanging-drop method. The spheroids were then loaded into a porous scaffold and induced to differentiate into cartilage. To confirm the cartilage phenotype of the differentiated spheroids in the scaffold, the complex was evaluated for the expression of chondrogenic-related genes and proteins. The cartilagenous tissues formed from the spheroid-scaffold complexes were primarily checked for in vivo functioning by transplanting them into rat models of cartilage damage. The results showed that the spheroid-scaffold complexes displayed the cartilage phenotype after inducing chondrogenic differentiation. The complexes stained positive with safranin O, alcian blue and collagen 2, significantly expressed Sox9, Col2, and aggrecan genes compared with that before differentiation. In vivo, the differentiated spheroid-scaffold complexes formed mature cartilage that stained strongly positive with safranin O and fast red. These results suggest a promising strategy to produce cartilaginous microtissue for regenerative medicine.


Subject(s)
Cartilage/cytology , Mesenchymal Stem Cells/cytology , Spheroids, Cellular/metabolism , Tissue Engineering/methods , Tissue Scaffolds , Cartilage/metabolism , Cell Differentiation , Humans , In Vitro Techniques , Mesenchymal Stem Cells/metabolism , Porosity
12.
Adv Exp Med Biol ; 1292: 145-155, 2020.
Article in English | MEDLINE | ID: mdl-32430853

ABSTRACT

INTRODUCTION: The breast cancer cells with CD44+CD24- phenotype are known to play an important role in tumorigenesis, drug resistance, and cancer recurrence. Breast cancer cells with CD44+CD24- phenotype are cultured in three-dimensional (3D) stereotype showing the recapitulation of tumors in vivo such as cell differentiation, heterogeneity, and microenvironment. Using this 3D model in anti-cancer compound research results in a more accurate reflection than conventional monolayer cell culture. This study aimed to identify the antitumor activity of Hopea odorata methanol extract (HO-MeOH-E) on breast cancer cells and cancer stem-like cells in both models of three-dimensional culture (3D) and monolayer cell culture (2D). METHODS: HO-MeOH-E was produced from Hopea odorata plant. The VN9 breast cancer cells (VN9) were collected and expanded from the previous study. The breast cancer stem-like cells (VN9CSC) were sorted from the VN9 based on phenotype CD44+CD24-. Both VN9 and VN9CSC were used to culture in monolayer culture (2D) and organoids (3D) before they were used to treat with HO-MeOH-E. Two other anticancer drugs, doxorubicin and tirapazamine, were used as references. The antitumor activities of extracts and drugs were determined via two assays: antiproliferation using the Alamar blue assay and cell cycle assay. RESULTS: The results showed that HO-MeOH-E was sensitive to both VN9 and VN9CSC in 3D more than 2D culture (IC50 on 3D organoids 144.8 ± 2.172 µg/mL and on 2D 340.2 ± 17.01 µg/mL for VN9CSC (p < 0.001); IC50 on 3D organoids 2055 ± 82.2 µg/mL and on 2D 430.6 ± 8.612 µg/mL for VN9 (p < 0.0001), respectively). HO-MeOH-E inhibits VN9CSC proliferation by blocking S phase and increasing the populations of apoptotic cells; this is consensus to the effect of tirapazamine (TPZ) which is used in hypoxia-activated chemotherapy. CONCLUSION: Taken these results, HO-MeOH-E has the potential effect in hypoxia-activated chemotherapy specifically on breast cancer stem-like cells with CD44+CD24- phenotype.


Subject(s)
Breast Neoplasms/pathology , Cell Culture Techniques , Cell Death/drug effects , Dipterocarpaceae/chemistry , Neoplastic Stem Cells/drug effects , Plant Extracts/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Humans , Neoplastic Stem Cells/pathology
13.
Adv Exp Med Biol ; 1292: 113-130, 2020.
Article in English | MEDLINE | ID: mdl-32430854

ABSTRACT

The idea of utilizing the human immune system to eradicate tumors has been successfully practiced for the past decades, as reported in multiple published studies. Among cancer types, non-small cell lung cancer (NSCLC) is considered the most lethal type, leading to the necessity of finding an effective treatment for this category of cancer. Building on the success of basic and preclinical studies, numerous clinical trials of cytokine-induced killer (CIK) cells or chimera antigen receptor (CAR) T cells for NSCLC therapy have been reported. In this review paper, we will summarize those findings in the context of clinical outcomes and adverse effects. In NSCLC, compared to CAR-T cells, CIK cells show relatively stronger antitumor efficacy and lower adverse effects. More clinical studies are needed to further confirm the clinical efficiency of both types of cellular immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Cytokine-Induced Killer Cells/immunology , Immunotherapy, Adoptive , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/transplantation , Cell Transplantation , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
Stem Cell Res Ther ; 11(1): 60, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054512

ABSTRACT

INTRODUCTION: Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide. COPD results from chronic inflammation of the lungs. Current treatments, including physical and chemical therapies, provide limited results. Stem cells, particularly mesenchymal stem cells (MSCs), are used to treat COPD. Here, we evaluated the safety and efficacy of umbilical cord-derived (UC)-MSCs for treating COPD. METHODS: Twenty patients were enrolled, 9 at stage C and 11 at stage D per the Global Initiative for Obstructive Lung Disease (GOLD) classification. Patients were infused with 106 cells/kg of expanded allogeneic UC-MSCs. All patients were followed for 6 months after the first infusion. The treatment end-point included a comprehensive safety evaluation, pulmonary function testing (PFT), and quality-of-life indicators including questionnaires, the 6-min walk test (6MWT), and systemic inflammation assessments. All patients completed the full infusion and 6-month follow-up. RESULTS: No infusion-related toxicities, deaths, or severe adverse events occurred that were deemed related to UC-MSC administration. The UC-MSC-transplanted patients showed a significantly reduced Modified Medical Research Council score, COPD assessment test, and number of exacerbations. However, the forced expiratory volume in 1 s, C-reactive protein, and 6MWT values were nonsignificantly reduced after treatment (1, 3, and 6 months) compared with those before the treatment. CONCLUSION: Systemic UC-MSC administration appears to be safe in patients with moderate-to-severe COPD, can significantly improve their quality of life, and provides a basis for subsequent cell therapy investigations. TRIAL REGISTRATION: ISRCTN, ISRCTN70443938. Registered 06 July 2019.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Pulmonary Disease, Chronic Obstructive/therapy , Transplantation, Homologous/methods , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Pilot Projects
15.
Adv Exp Med Biol ; 1292: 13-25, 2020.
Article in English | MEDLINE | ID: mdl-29687285

ABSTRACT

BACKGROUND: Cancer is one of the leading causes of death in the world. A great deal of effort has been made to discover new agents for cancer treatment. Xao tam phan (Paramignya trimera) is a traditional medicine of Vietnam used in cancer treatment for a long time, yet there is not much scientific evidence proving its anticancer potency. The study aimed to evaluate the toxicity of Paramignya trimera extract (PTE) on multicellular tumor spheres (MCTS) of MCF-7 cells using hanging drop technique. METHODS: Firstly, MCF-7 cells were seeded on hanging drop plates, spheroid size was tracked, and growth curve was measured by MTT assay and AlamarBlue® assay. The necrotic core of MCTS was evaluated by propidium iodide (PI) staining. Toxicity of doxorubicin (DOX) and tirapazamine (TPZ) was then tested on 3D model compared to 2D culture condition. RESULTS: The results showed that the IC50 of DOX on 3D MCF-7 cells was nearly 50 times greater than monolayer MCF-7 cells. In contrast, TPZ (an agent which is specifically toxic under hypoxic conditions) had significantly lower IC50 in 3D condition than in 2D. The toxicity tests for PTE showed that PTE strongly inhibited MCF-7 cells in both 2D and 3D conditions. Interestingly, the IC50 of PTE in 3D model was remarkably lower than in 2D (IC50 value was 168.9 ± 11.65 µg/ml compared to 260.8 ± 16.54 µg/ml, respectively). The invasion assay showed that PTE completely inhibited invasion of MCF-7 cells at 250 µg/mL concentration. Also, flow cytometry results indicated that PTE effectively induced apoptosis in MCF-7 spheroids in 3D condition at 250 µg/mL concentration. CONCLUSION: The results from this study emphasize the promise of PTE in cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Cell Culture Techniques/methods , Methanol/chemistry , Models, Biological , Plant Extracts/chemistry , Plant Extracts/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/toxicity , Humans , MCF-7 Cells
16.
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
17.
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
18.
Adv Exp Med Biol ; 1084: 109-128, 2019.
Article in English | MEDLINE | ID: mdl-30242785

ABSTRACT

INTRODUCTION: Since the 1980s, adipose-derived stem cells (ASCs) have become a powerful and potential source for stem cell-based therapy, regenerative medicine, and even drug delivery in cancer treatment. The development of off-the-shelf mesenchymal stem cells (MSCs), including ASCs, has rapidly advanced in recent years with several clinical trials and approved products. In this technology, ASCs should be expanded long term in order to harvest higher cell number. In this study, senescence of ASCs after long-term expansion was evaluated. METHODS: Human ASCs (hASCs) were isolated and cultured continuously at a density of 103 cells/cm2 up to passage 15. The cells were assessed for aging via changes in the following: characteristics of MSCs, mitochondrial activity, accumulation of beta-galactosidase, and expression of tumor suppressor genes. RESULTS: The results showed that following in vitro expansion to the 15th passage, ASCs did not show changes in immunophenotype, except for decreased expression of CD105. However, the cells increased in size and in shape and complexity (toward the "fried egg" morphology). They also almost ceased to proliferate in passage 15. Nonetheless, they maintained in vitro differentiation potential toward osteoblasts, chondrocytes, and adipocytes. Expression of tumor suppressor genes p53 and p16 did not significantly change, while p27 was significantly downregulated. Mitochondrial activities also decreased slightly in culture from passage 5 to passage 10 and remained stable to passage 15. ASCs also showed increased accumulation of beta-galactosidase in culture, but it was negligible. CONCLUSION: In conclusion, hASCs exhibited some particular characteristics of aged stem cells when the number of subculture cells increased. However, up to passage 10, ASCs also retained almost all of the characteristics of MSCs.


Subject(s)
Adipocytes , Adipose Tissue , Cellular Senescence , Mesenchymal Stem Cells , Adipocytes/cytology , Adipose Tissue/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Stem Cells
19.
Adv Exp Med Biol ; 1083: 145-156, 2018.
Article in English | MEDLINE | ID: mdl-29423674

ABSTRACT

Mesenchymal stem cell (MSC) transplantation is a novel treatment for diabetes mellitus, especially type 1 diabetes. Many recent publications have demonstrated the efficacy of MSC transplantation on reducing blood glucose and increasing insulin production in both preclinical and clinical trials. However, the investigation of grafted cell doses has been lacking. Therefore, this study aimed to evaluate the different doses of MSCs on treatment of type 1 diabetes in mouse models. MSCs were isolated and expanded from human adipose tissue. Streptozotocin (STZ)-induced diabetic mice were divided into two groups that were intravenously transfused with two different doses of human MSCs: 106 or 2.106 cells/mouse. After transplantation, both grafted and placebo mice were monitored weekly for their blood glucose levels, glucose and insulin tolerance, pancreatic structural changes, and insulin production for 56 days after transplantation. The results showed that the higher dose of MSCs (2.106 cells/mouse) remarkably reduced death rate. The death rates were 50%, 66%, and 0% in placebo group, low-dose (1.106 MSCs) group, and high-dose (2.106 MSCs) group, respectively, after 56 days of treatment. Moreover, blood glucose levels were lower for the high-dose group compared to other groups. Glucose and insulin tolerance, as well as insulin production, were significantly improved in mice transplanted with 2.106 cells. The histochemical analyses also support these results. Thus, a higher (e.g., 2.106) dose of MSCs may be an effective dose for treatment of type 1 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Mesenchymal Stem Cell Transplantation , Adipose Tissue/cytology , Animals , Blood Glucose , Glucose Tolerance Test , Humans , Infusions, Intravenous , Insulin/blood , Mesenchymal Stem Cells/cytology , Mice
20.
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.

SELECTION OF CITATIONS
SEARCH DETAIL
...