Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Cancer Res Clin Oncol ; 150(6): 299, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38850382

ABSTRACT

BACKGROUND: Microvesicles are membraned particles produced by different types of cells recently investigated for anticancer purposes. The current study aimed to investigate the effects of human bone marrow mesenchymal stem cell-derived microvesicles (BMSC-MVs) on the multiple myeloma cell line U266. BMSC-MVs were isolated from BMSCs via ultracentrifugation and characterized using transmission electron microscopy (TEM) and dynamic light scattering (DLS). U266 cells were treated with 15, 30, 60, and 120 µg/mL BMSC-MVs for three and seven days and the effects of treatment in terms of viability, cytotoxicity, and DNA damage were investigated via the MTT assay, lactate dehydrogenase (LDH) assay, and 8­hydroxy-2'-deoxyguanosine (8­OHdG) measurement, respectively. Moreover, the apoptosis rate of the U266 cells treated with 60 µg/mL BMSC-MVs was also assessed seven days following treatment via flow cytometry. Ultimately, the expression level of BCL2, BAX, and CCND1 by the U266 cells was examined seven days following treatment with 60 µg/mL BMSC-MVs using qRT-PCR. RESULTS: BMSC-MVs had an average size of ~ 410 nm. According to the MTT and LDH assays, BMSC-MV treatment reduced the U266 cell viability and mediated cytotoxic effects against them, respectively. Moreover, elevated 8­OHdG levels following BMSC-MV treatment demonstrated a dose-dependent increase of DNA damage in the treated cells. BMSC-MV-treated U266 cells also exhibited an increased apoptosis rate after seven days of treatment. The expression level of BCL2 and CCND1 decreased in the treated cells whereas the BAX expression demonstrated an incremental pattern. CONCLUSIONS: Our findings accentuate the therapeutic benefit of BMSC-MVs against the multiple myeloma cell line U266 and demonstrate how microvesicles could be of therapeutic advantage. Future in vivo studies could further corroborate these findings.


Subject(s)
Apoptosis , Cell-Derived Microparticles , Mesenchymal Stem Cells , Multiple Myeloma , Humans , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Mesenchymal Stem Cells/metabolism , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Cell Survival , DNA Damage
2.
Mol Neurobiol ; 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38010560

ABSTRACT

Memory problems are often the first signs of cognitive impairment related to Alzheimer's disease (AD), and stem cells and stem cell-derived exosomes (EXOs) have been studied for their therapeutic potential to improve the disease signs. While many studies have shown the anti-inflammatory and immunomodulatory effects of stem cells and exosomes on improving memory in different AD models, there is still insufficient data to determine how they modulate neural plasticity to enhance spatial memory and learning ability. Therefore, we conducted a study to investigate the effects of exosomes derived from 3D-cultured human Unrestricted Somatic Stem Cells (hUSSCs) on spatial memory and neuroplasticity markers in a sporadic rat model of AD. Using male Wistar rats induced by intracerebral ventricle injection of streptozotocin, we demonstrated that intranasal administration of hUSSC-derived exosomes could decrease Aß accumulation and improve learning and memory in the Morris water maze test. We also observed an increase in the expression of pre-synaptic and post-synaptic molecules involved in neuronal plasticity, including NMDAR1, integrin ß1, synaptophysin, pPKCα, and GAP-43, in the hippocampus. Our findings suggest that intranasal administration of exosomes can ameliorate spatial learning and memory deficits in rats, at least in part, by increasing the expression of neuroplasticity proteins. These results may encourage researchers to further investigate the molecular pathways involved in memory improvement after stem cell and exosome therapy, with the goal of increasing the efficacy and safety of exosome-based treatments for AD.

3.
Stem Cells Int ; 2023: 8836452, 2023.
Article in English | MEDLINE | ID: mdl-37576406

ABSTRACT

Background: Although several studies have been conducted on modeling human liver disease, it is still challenging to mimic nonalcoholic fatty liver disease in vitro. Here, we aimed to develop a fibrotic liver microtissue composed of hepatocytes, hepatic stellate, and endothelial cells. In addition, the therapeutic effects of umbilical cord mesenchymal stem cell-derived exosomes (UC-MSC-EXO) and anti-miR17-5p as new antifibrotic drugs were investigated. Methods: To create an effective preclinical fibrosis model, multicellular liver microtissues (MLMs) consisting of HepG2, LX2, and HUVECs were cultured and supplemented with a mixture of palmitic acid and oleic acid for 96 hr. Then, MLMs were exposed to UC-MSC-EXO and anti-miR17-5p in different groups. The results of cell viability, reactive oxygen species (ROS) production, liver enzyme levels, inflammation, and histopathology were analyzed to assess the treatment efficacy. Furthermore, the expression of collagen I (COL I) and α-smooth muscle actin (α-SMA) as critical matrix components, transforming growth factor beta (TGF-ß), and miR-17-5p were measured. Results: Free fatty acid supplementation causes fibrosis in MLMs. Our results demonstrated that UC-MSC-EXO and anti-miR17-5p attenuated TGF-ß1, interleukin-1ß, and interleukin-6 in all experimental groups. According to the suppression of the TGF-ß1 pathway, LX2 activation was inhibited, reducing extracellular matrix proteins, including COL I and α-SMA. Also, miR-17-5p expression was elevated in fibrosis conditions. Furthermore, we showed that our treatments decreased alanine aminotransferase and aspartate aminotransferase, and increased albumin levels in the culture supernatant. We also found that both MSC-EXO and MSC-EXO + anti-miR17-5p treatments could reduce ROS production. Conclusion: Our findings indicated that anti-miR17-5p and MSC-EXO might be promising therapeutic options for treating liver fibrosis. Furthermore, EXO + anti-miR had the best effects on boosting the fibrotic markers. Therefore, we propose this novel MLM model to understand fibrosis mechanisms better and develop new drugs.

4.
Noncoding RNA Res ; 8(3): 451-458, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37455764

ABSTRACT

Background: Anaplastic thyroid cancer (ATC) is one of the most aggressive malignancies in humans that accounts for a considerable rate of cancer-associated mortality. Since conventional therapies are lacking sufficient efficacy, new treatment approaches are required. This goal could be achieved through a better understanding of the molecular pathogenesis of ATC. Thyroid tumorigenesis is initiated by a subpopulation of cells known as cancer stem cells (CSCs) with specific markers such as CD133 that confers to processes such as self-renewal and metastasis. Besides, some long non-coding RNAs (lncRNAs) promote tumorigenesis by mediating the aforementioned processes. Methods: Here, we designed an exploratory study to investigate the role of lncRNAs ROR and MALAT1 and their related genes in CSC stemness. Using magnetic-activated cell sorting (MACS), the CD133- and CD133+ subpopulations were separated in SW1736 and C643 ATC cell lines. Next, the expression profiles of the CD133 marker, MALAT1, and its associated genes (CCND1, NESTIN, MYBL2, MCL1, IQGAP1), as well as ROR and its related genes (POU5F1, SOX2, NANOG), were explored by qRT-PCR. Results: We found significant up-regulation of ROR, POU5F1, SOX2, NANOG, CD133, MALAT1, IQGAP1, and MCL1 in CD133+ SW1736 cells compared to CD133- cells. As for CD133+ C643 cells, CCND1, IQGAP1, POU5F1, SOX2, NANOG, and NESTIN were significantly up-regulated compared to CD133- cells. Conclusions: This study suggests that these lncRNAs in CD133-positive SW1736 and C643 cells might regulate stemness behaviors in ATC.

5.
APMIS ; 131(8): 381-393, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36867046

ABSTRACT

It has long been hypothesized that leukemic cells are able to modulate the fate of resident cells in the tumor microenvironment (TME) toward either supporting or immunosuppressive cells for the development of tumors. Exosomes can be a potential culprit in imposing tumor desire. There is evidence about the impact of tumor-derived exosomes on different immune cells in different malignancies. However, findings about macrophages are contradictory. Here, we evaluated the potential influence of multiple myeloma (MM)-cell-derived exosomes on the polarization of macrophages by examining hallmarks of M1 and M2 macrophages. After treatment of M0 macrophages with isolated exosomes (from U266B1), gene expression (Arg-1, IL-10, TNF-α and IL-6), immunophenotyping markers (CD206), cytokine secretion (IL-10 and IL-6), nitric oxide (NO) production, and redox potentiality of target cells were assessed. Our results revealed significantly increased expression of the genes involved in the development of M2-like cells but not M1 cells. The CD 206 marker and IL-10 protein levels were significantly increased at different time points. The expression of IL-6 mRNA and IL-6 protein secretion did not change significantly. MM-cell-derived exosomes induced significant changes in NO production and intracellular ROS levels in M0 cells.


Subject(s)
Exosomes , Multiple Myeloma , Humans , Exosomes/genetics , Exosomes/metabolism , Interleukin-10/genetics , Interleukin-6 , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Macrophages , Tumor Microenvironment
6.
Exp Gerontol ; 175: 112142, 2023 05.
Article in English | MEDLINE | ID: mdl-36921675

ABSTRACT

Blood donor age has become a major concern due to the age-associated variations in the content and concentration of circulating extracellular nano-sized vesicles (EVs), including exosomes. These EVs mirror the state of their parental cells and transfer it to the recipient cells via biological messengers such as microRNAs (miRNAs, miRs). Since the behavior of hematopoietic stem cells (HSCs) is potentially affected by the miRs of plasma-derived EVs, a better understanding of the content of EVs is important for the safety and efficacy perspectives in blood transfusion medicine. Herein, we investigated whether the plasma-derived EVs of young (18-25 years) and elderly human donors (45-60 years) can deliver "youth" or "aging" signals into human umbilical cord blood (hUCB)-derived HSCs in vitro. The results showed that EVs altered the growth functionality and differentiation of HSCs depending on the age of the donor from which they are derived. EVs of young donors could ameliorate the proliferation and self-renewal potential of HSCs whereas those of aged donors induced senescence-associated differentiation in the target cells, particularly toward the myeloid lineage. These findings were confirmed by flow cytometric analysis of surface markers and microarray profiling of genes related to stemness (e.g., SOX-1, Nanog) and differentiation (e.g., PU-1). The results displayed an up-regulation of miR-29 and miR-96 and a down-regulation of miR-146 in EVs derived from elderly donors. The higher expression of miR-29 and miR-96 contributed to the diminished expression of CDK-6 and CDKN1A (p21), promoting senescence fate via cell growth suppression, while the lower expression of miR-146 positively regulates TRAF-6 expression to accelerate biological aging. Our findings reveal that plasma-derived EVs from young donors can reverse the aging-associated changes in HSCs, while vice versa, the EVs from elderly donors rather promote the senescence process.


Subject(s)
Extracellular Vesicles , MicroRNAs , Aged , Humans , Rejuvenation , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/metabolism , Cell Differentiation , Hematopoietic Stem Cells
7.
Biochimie ; 209: 73-84, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36681232

ABSTRACT

Skin wound healing is a multifaceted process involving a cascade of molecular and cellular procedures that occur in four different phases: (a) hemostasis, (b) inflammation, (c) proliferation, and (d) tissue remodeling. Prolonged wound healing in skin is still a major challenge in treatment of wounds. Mesenchymal stem cells (MSCs) accelerate cutaneous wound healing through their paracrine activity. Exosomes are one of the key secretory products of MSCs, mimicking the effects of parental MSCs in skin wound healing process. Exosomes are small membrane vesicles (30-150 nm in diameter) that originate from endosomal pathways and transport numerous biomolecules, including DNAs, messenger RNAs, microRNAs, lipids, and proteins. They can be taken up by target cells and release their contents to modulate the activity of recipient cells. Exosomes derived from mesenchymal stem cells (MSC-Exo) reduce inflammation, promote proliferation, inhibit apoptosis, and enhance angiogenesis in skin wound healing process. Therefore, exosomes are emerging as novel cell-cell communication mediators and have opened a novel viewpoint for developing cell-free therapies. This review aims to demonstrate the roles of exosomes in each step of skin wound healing through a comprehensive literature search.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Exosomes/metabolism , Wound Healing , Skin/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism
8.
Cells Tissues Organs ; 212(2): 164-175, 2023.
Article in English | MEDLINE | ID: mdl-34749364

ABSTRACT

Sepsis is a systemic infection mainly caused by bacterial infections. Despite all efforts and advances in the treatment of sepsis, it is still considered one of the leading causes of death in hospitalized patients. Today, we have to use novel therapies and one of the most important is cell-free therapy. Exosomes have been shown to contain the contents of their parent cells and that they do not generate an immune response between different individuals which makes them a good candidate for transplantation. Unrestricted somatic stem cells (USSC), also known as mesenchymal stem cell progenitors due to their high proliferative capacity and low immune response, may be a novel therapy for sepsis. In this study, the effect of USSC-derived exosomes on sepsis was investigated using a mouse model. USSCs were isolated from human cord blood and characterized by flow cytometry and multi-lineage differentiation. The exosomes were then harvested from USSCs and characterized by transmission electron microscopy, Western blotting, and dynamic light scattering. The harvested exosomes were injected into the mouse model of sepsis. Biochemical, histological, molecular, and survival studies were performed in different groups. Our observations showed that USSC-derived exosomes can reduce inflammation in septic mice. Histopathologic and biochemical findings in the sham group showed multiorgan involvement, but these changes disappeared after 7 days of exosome administration. Moreover, the expression of IRAK-1 and TRAF-6 (main adapter molecules in signaling pathways of inflammation) was decreased through negative regulation by miR-146a after 72 h of exosome administration. A 2-fold increase in the level of IL-10 and a 2-fold decrease in the levels of IL-6 and TNF-α was observed. In conclusion, we showed that direct injection of USSC-derived exosomes can be one of the important methods for the treatment of various aspects of sepsis due to their immunomodulatory properties.


Subject(s)
Adult Stem Cells , Exosomes , Sepsis , Animals , Mice , Humans , Disease Models, Animal , Exosomes/metabolism , Inflammation/metabolism , Sepsis/therapy
9.
Cell J ; 24(11): 689-696, 2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36377219

ABSTRACT

OBJECTIVE: Angiogenesis has critical roles in several physiological processes. Restoring angiogenesis in some pathological conditions such as a few vascular diseases can be a therapeutic approach to controlling this issue. Mesenchymal stem cells (MSCs) secrete specific intracellular products known as extracellular vesicles (EVs) with high therapeutic potential which compared to their source cells, do not have the limitations of cell therapy. The angiogenic effect of the human umbilical cord MSCs (hUCMSCs)-derived small EVs are evaluated in the present work. Aim of this research is to show that hUCMSCs-derived small EVs cause differentiation of genes involved in angiogenesis like FGFR-1, FGF, VEGF, and VEGFR-2. MATERIALS AND METHODS: In this experimental study, MSCs were isolated from the human umbilical cord, and after confirming their identities, their secreted EVs (including exosomes) were extracted by ultracentrifugation. The isolated small EVs were characterized by dynamic light scattering (DLS), transmission electron microscopy (TEM), bicinchoninic acid assay (BCA), and Western Blotting. Then, the human umbilical vein endothelial cells (HUVECs) were treated with derived small EVs for 72 hours, and the expression of the angiogenic factors including FGFR-1, FGF, VEGF, and VEGFR-2 was evaluated by quantitative real-time-polymerase chain reaction (qPCR). Angiogenesis was also evaluated via a tube formation assay. RESULTS: The results demonstrated that FGFR-1, FGF, VEGF, and VEGFR-2 could be elevated 2, 2, 3.5, and 2 times, respectively, in EVs treated HUVECs, and derivative EVs can encourage tube formation in HUVECs. CONCLUSION: These findings imply that hUCMSCs-derived small EVs are valuable resources in promoting angiogenesis and are very promising in cell-free therapy.

10.
Stem Cell Res Ther ; 13(1): 194, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35550188

ABSTRACT

Exosomes are extracellular vesicles found in various tissues, blood circulation, and tissue fluids, secreted into the extracellular environment by fusing a multivesicular body with a plasma membrane. Various cell types release these vesicles to contribute to many cellular functions, including intercellular communication, cell proliferation, differentiation, angiogenesis, response to stress, and immune system signaling. These natural nanoparticles have therapeutic effects in various diseases and exhibit a behavior similar to the cell from which they originated. In the meantime, exosomes derived from mesenchymal stem cells have attracted the attention of many researchers and physicians due to their unique ability to modulate the immune system, repair tissue and reduce inflammation. Numerous clinical and preclinical studies have examined the effect of MSC-derived exosomes in various diseases, and their results have been published in prestigious journals. This review article discusses the biogenesis and sources of exosomes, MSC-derived exosomes, the use of these exosomes in regenerative medicine, and treatments based on exosomes derived from stem cells in respiratory diseases.


Subject(s)
Exosomes , Extracellular Vesicles , Mesenchymal Stem Cells , Respiratory Tract Diseases , Cell Differentiation , Exosomes/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Regenerative Medicine/methods , Respiratory Tract Diseases/metabolism , Respiratory Tract Diseases/therapy
11.
Stem Cell Res Ther ; 12(1): 91, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514427

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a fatal complication of coronavirus disease 2019 (COVID-19). There are a few reports of allogeneic human mesenchymal stem cells (MSCs) as a potential treatment for ARDS. In this phase 1 clinical trial, we present the safety, feasibility, and tolerability of the multiple infusions of high dose MSCs, which originated from the placenta and umbilical cord, in critically ill COVID-19-induced ARDS patients. METHODS: A total of 11 patients diagnosed with COVID-19-induced ARDS who were admitted to the intensive care units (ICUs) of two hospitals enrolled in this study. The patients were critically ill with severe hypoxemia and required mechanical ventilation. The patients received three intravenous infusions (200 × 106 cells) every other day for a total of 600 × 106 human umbilical cord MSCs (UC-MSCs; 6 cases) or placental MSCs (PL-MSCs; 5 cases). FINDINGS: There were eight men and three women who were 42 to 66 years of age. Of these, six (55%) patients had comorbidities of diabetes, hypertension, chronic lymphocytic leukemia (CLL), and cardiomyopathy (CMP). There were no serious adverse events reported 24-48 h after the cell infusions. We observed reduced dyspnea and increased SpO2 within 48-96 h after the first infusion in seven patients. Of these seven patients, five were discharged from the ICU within 2-7 days (average: 4 days), one patient who had signs of acute renal and hepatic failure was discharged from the ICU on day 18, and the last patient suddenly developed cardiac arrest on day 7 of the cell infusion. Significant reductions in serum levels of tumor necrosis factor-alpha (TNF-α; P < 0.01), IL-8 (P < 0.05), and C-reactive protein (CRP) (P < 0.01) were seen in all six survivors. IL-6 levels decreased in five (P = 0.06) patients and interferon gamma (IFN-γ) levels decreased in four (P = 0.14) patients. Four patients who had signs of multi-organ failure or sepsis died in 5-19 days (average: 10 days) after the first MSC infusion. A low percentage of lymphocytes (< 10%) and leukocytosis were associated with poor outcome (P = 0.02). All six survivors were well with no complaints of dyspnea on day 60 post-infusion. Radiological parameters of the lung computed tomography (CT) scans showed remarkable signs of recovery. INTERPRETATION: We suggest that multiple infusions of high dose allogeneic prenatal MSCs are safe and can rapidly improve respiratory distress and reduce inflammatory biomarkers in some critically ill COVID-19-induced ARDS cases. Patients that develop sepsis or multi-organ failure may not be good candidates for stem cell therapy. Large randomized multicenter clinical trials are needed to discern the exact therapeutic potentials of MSC in COVID-19-induced ARDS.


Subject(s)
COVID-19/therapy , Mesenchymal Stem Cell Transplantation , Respiratory Distress Syndrome/therapy , Adult , Aged , Biomarkers/blood , Comorbidity , Critical Care , Critical Illness , Female , Humans , Hypoxia/virology , Inflammation , Intensive Care Units , Lung/diagnostic imaging , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Patient Safety , Placenta/cytology , Pregnancy , Respiration, Artificial , Respiratory Distress Syndrome/virology , Sepsis/virology , Tomography, X-Ray Computed , Transplantation, Homologous , Treatment Outcome , Umbilical Cord/cytology
12.
J Biomater Appl ; 35(10): 1315-1326, 2021 05.
Article in English | MEDLINE | ID: mdl-33307942

ABSTRACT

Functional cartilage tissue engineering needs a substantial, easy to handle scaffold with proper mechanical strength to repair defected area in articular cartilage. In this study, we report the development and characterization of demineralized bone matrix (DBM) in with a poly vinyl alcohol (PVA) to have a proper homogenous injectable scaffold. Injectabiliy of the biodegradable scaffolds, degradation rate, swelling ratio compression and tensile mechanical properties, and viability and proliferation of bone marrow mesenchymal stem cells (BM-MSCs) followed by differentiation of them In-vitro and In-vivo seeded within the scaffold were studied. It demonstrated that the PVA 20% could increase significantly (p < 0.05) the biodegradability of DBM after 720 hours.DBM with 20% of PVA scaffold has significantly higher (p < 0.05) compression and tensile mechanical strength and viscosity. SEM images showed a multilayer of cells on DBM scaffold incorporated with PVA 20%.BM-MSCs on scaffolds, DBM+PVA 20% had a significant growth rate (p < 0.0001) compare to 2D and low concentration of PVA after 21 days of culture. Viability of cells was significantly higher (p < 0.05) on DBM+PVA scaffold compare to DBM. DBM+PVA 20% enhanced cell viability (P < 0.05) compare to DBM scaffold. The PVA presence enhanced chondrogenesis differentiation at the cellular and molecular levels, as evidenced by increased COL II (P < 0.05) and SOX2 upregulation of Chondrogensis-specific genes (p < 0.001). Hyline-like cartilage covered the defect which was confirmed by microscopy and histology assessments. Having considered percentages of PVA with a constant amount of DBM, injectability, compressive mechanical properties, homogeneity of the scaffold, and providing sufficient surface area (12.25 cm2/ml) for cell attachment; 0.35 g/ml of DBM in 20% PVA (w/v) has applicable properties within the ranges of studies which can be proposed for the injectable engineered articular cartilage.


Subject(s)
Biocompatible Materials/chemistry , Bone Matrix/chemistry , Polyvinyl Alcohol/chemistry , Animals , Biocompatible Materials/pharmacology , Cartilage, Articular/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chondrogenesis/drug effects , Collagen Type II/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Models, Animal , Prostheses and Implants , Rabbits , SOXB1 Transcription Factors/metabolism , Tissue Engineering , Tissue Scaffolds , Up-Regulation/drug effects
13.
Am J Blood Res ; 10(5): 217-230, 2020.
Article in English | MEDLINE | ID: mdl-33224566

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) represents a vital curative choice for many disease. However its outcome can be hampered by a variety of transplant associated complications. Hemorrhagic cystitis (HC) considered as one of the major difficulties after HSCT. HC symptoms comprise hematuria, dysuria, burning during urination, urinary frequency, urgency and incontinency, abdominal or suprapubic pain, urinary obstruction, and renal or bladder damage. There are a lot of causes for HC development. BK virus reactivation is one of the major causes of HC after HSCT. There is still no standard and approved treatment protocol for BK virus associated HC (BKV-HC). Treatment of HC is according to the local standard operating procedures, depending on the cause and severity. In this study we will review the current treatments available for this disease. We have divided the therapeutic procedures into 5 categories including conservative therapy, complimentary options, surgical procedures, pharmacological treatments and adoptive cell therapy. We believe that comparing the advantages and disadvantages of different therapies make it easier to choose the best treatment protocol. In addition, we had a greater focus on adoptive cell therapy, because it is a relatively new introduced method and might be a logical alternative to conventional treatments for refractory patients. In total, no definitive recommendation is possible for current available treatments because these procedures have only been utilized sporadically in a limit number of patients. Furthermore, a number of treatment options are only experimental and definitely need more effort.

14.
Mater Sci Eng C Mater Biol Appl ; 104: 109921, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31500009

ABSTRACT

Graphene based nanomaterials are promising candidates for cardiac tissue engineering due to the excellent electrical and mechanical properties and the robust surface chemistry. This research was designed to investigate the physicochemical and biological effects of increasing concentration of reduced graphene oxide (rGO) coating on collagen (Col) scaffolds as well as their antibacterial properties. Enhanced GO coating content to 400 µg/ml and its reduction showed improvement of HUVECs viability, however, following reduction of more GO concentration, decreased cell viability was observed. Compared with the Col counterpart, electroactive containing rGO scaffolds upregulated cardiac gene expression involve in electrical coupling (Cx43), muscle contraction and relaxation (troponin-T) and cytoskeleton alignment (actinin-4) after 7 days even without external electrical stimulation. rGO coating significantly improved mechanical properties and the electroactivity of the Col scaffolds reaching to 1100 ±â€¯31 kPa and 4 × 10-4 ±â€¯1.20 S/m for GO concentration of 800 µg/ml, respectively. Also, the antibacterial properties of Col-rGO-400 scaffolds against Escherichia coli, Staphylococcus aureus and Streptococcus pyogenes were confirmed by culture and FESEM observation. Taken together, the results indicated that rGO coating presents promising properties to Col scaffolds providing a desirable micro environment for cardiomyocytes coupling and gene upregulation as well as antibacterial activities for cardiac patch application.


Subject(s)
Anti-Bacterial Agents/pharmacology , Electricity , Graphite/pharmacology , Myocardium/cytology , Tissue Scaffolds/chemistry , Bacteria/drug effects , Bacteria/growth & development , Bacterial Adhesion/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Humans , Microbial Sensitivity Tests , Oxidation-Reduction , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
15.
J Cell Biochem ; 120(7): 11441-11453, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30746766

ABSTRACT

The function of fibroblast cells in wounded areas results in reconstruction of the extra cellular matrix and consequently resolution of granulation tissue. It is suggested that the use of platelet-rich plasma can accelerate the healing process in nonhealing or slow-healing wounds. In this study, a simple and novel method has been used to fabricate an electrospun three-layered scaffold containing plasma rich in growth factor with the aim of increasing the proliferation and migration of fibroblast cells in vitro. First, plasma rich in growth factor was derived from platelet rich plasma, and then a three-layered scaffold was fabricated using PLLA nanofibers as the outer layers and plasma rich in growth factor-containing gelatin fibers as the internal layer. The growth morphology of cells seeded on this scaffold was compared to those seeded on one layered PLLA scaffold. The study of the cell growth rate on different substrates and the migration of cells in response to the drug release of multilayered scaffold was investigated by the cell quantification assay and a modified under agarose assay. Scanning electron microscopy and fluorescence images showed that cells seeded on multilayered scaffold were completely oriented 72 hours after seeding compared to those seeded on PLLA scaffold. The cell quantification assay also indicated significant increase in proliferation rate of cells seeded on three-layered scaffold compared to those seeded on PLLA scaffold and finally, monitoring cell migration proved that cells migrate significantly toward the three-layered scaffold up to 48 to 72 hours and afterwards start to show a diminished migration rate toward this scaffold.

16.
Tumour Biol ; 37(7): 9527-34, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26790441

ABSTRACT

The sprouting of new blood vessels by angiogenesis is critical in vascular development and homeostasis. Aberrant angiogenesis leads to enormous pathological conditions such as ischemia and cancer. MicroRNAs (also known as miRNAs or miRs) play key roles in regulation of a range of cellular processes by posttranscriptional suppression of their target genes. Recently, new studies have indicated that miRNAs are involved in certain angiogenic settings and signaling pathways use these non-coding RNAs to promote or suppress angiogenic processes. Herein, VEGFR2 and FGFR1 were identified as miR-129-1 and miR-133 targets using bioinformatic algorithms, respectively. Afterwards, using luciferase reporter assay and gene expression analysis at both mRNA and protein levels, VEGFR2 and FGFR1 were validated as miR-129-1 and miR-133 targets. In addition, we showed that miR-129-1 and miR-133 suppress angiogenesis properties such as proliferation rate, cell viability, and migration activity of human umbilical vein endothelial cells (HUVEC) in vitro. We conclude that these miRNAs can suppress key factors of angiogenesis by directly targeting them. These results have important therapeutic implications for a variety of diseases involving deregulation of angiogenesis, including cancer.


Subject(s)
Human Umbilical Vein Endothelial Cells/pathology , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Gene Expression/genetics , Humans , Neovascularization, Pathologic/pathology , RNA, Messenger/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
17.
J Med Genet ; 53(1): 24-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26510428

ABSTRACT

BACKGROUND: MicroRNA-129-1 (miR-129-1) seems to behave as a tumour suppressor since its decreased expression is associated with different tumours such as glioblastoma multiforme (GBM). GBM is the most common form of brain tumours originating from glial cells. The impact of miR-129-1 downregulation on GBM pathogenesis has yet to be elucidated. METHODS: MiR-129-1 was overexpressed in GBM cells, and its effect on proliferation was investigated by cell cycle assay. MiR-129-1 predicted targets (CDK6, IGF1, HDAC2, IGF2BP3 and MAPK1) were also evaluated by western blot and luciferase assay. RESULTS: Restoration of miR-129-1 reduced cell proliferation and induced G1 accumulation, significantly. Several functional assays confirmed IGF2BP3, MAPK1 and CDK6 as targets of miR-129-1. Despite the fact that IGF1 expression can be suppressed by miR-129-1, through 3'-untranslated region complementary sequence, we could not find any association between IGF1 expression and GBM. MiR-129-1 expression inversely correlates with CDK6, IGF2BP3 and MAPK1 in primary clinical samples. CONCLUSION: This is the first study to propose miR129-1 as a negative regulator of IGF2BP3 and MAPK1 and also a cell cycle arrest inducer in GBM cells. Our data suggests miR-129-1 as a potential tumour suppressor and presents a rationale for the use of miR-129-1 as a novel strategy to improve treatment response in GBM.


Subject(s)
Brain Neoplasms/genetics , Cell Cycle Checkpoints/genetics , Genes, Tumor Suppressor , Glioblastoma/genetics , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/genetics , RNA-Binding Proteins/genetics , Apoptosis/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Computational Biology , Cyclin-Dependent Kinase 6/genetics , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor I/genetics , MicroRNAs/chemistry , Mitogen-Activated Protein Kinase 1/chemistry , Models, Biological , RNA Interference , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry
18.
Appl Biochem Biotechnol ; 172(4): 2055-69, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24326679

ABSTRACT

Micro RNAs (miRNAs) are a novel class of non-coding regulatory RNA molecules that contribute to post-transcriptional gene regulation. Recent studies have demonstrated that specific miRNAs such as miR-150, miR-154, and miR-451 have key roles in erythropoiesis. To date, stimulatory cytokines are considered as unique effectors for in vitro differentiation of HSCs to erythropoietic lineage. However, the use of these factors is not cost-effective for clinical applications and therapeutic strategies. Here, we present a novel and cost-effective strategy in which miRNAs expression modulation promotes erythroid differentiation in HSCs in the absence of any extrinsic factors. Thus, CD133(+) hematopoietic stem cells purified from human umbilical cord blood were treated with pre-miR-451 containing lentiviruses, anti-miR-150 and anti-miR-154 in the absence of growth factors and cytokines. Obtained results indicated that miR-451 upregulation and miR-150 downregulation have positive effect on GATA-1, FOG-1, and EKLF, CD71 and CD235a genes expression and induce hemoglobinization efficiently. However, downregulation of miR-154 had no effect on erythropoiesis indexes compared to that observed in the control group. In conclusion, the data presented here for the first time demonstrate that expression modulation of miR-451 and miR-150 could be an efficient alternative to stimulatory cytokines for CD133(+) differentiation into erythroid lineage. Modulation of erythropoiesis in stem cells via miRNA holds promising potential for vascular tissue engineering and regenerative medicine applications.


Subject(s)
Erythroid Cells/cytology , Erythroid Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Female , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , MicroRNAs/physiology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...