Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Cardiol ; 410: 132227, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38844091

ABSTRACT

BACKGROUND: Acute ST-elevation myocardial infarction (STEMI) remains a globally significant health challenge in spite of improvement in management strategy. Being aware that mitochondrial dysfunction plays a crucial role in ischaemia-reperfusion injury (IRI) modulation, empirical evidence suggests functional mitochondrial transplantation strikes as a reliable therapeutic approach for patients with acute myocardial infarction. METHODS AND RESULTS: We conducted a prospective, triple-blinded, parallel-group, blocked randomised clinical trial to investigate the therapeutic effects and clinical outcomes of platelet-derived mitochondrial transplantation in 30 patients with acute STEMI, such that the 15 subjects in the control group were given standard of care treatment, whereas the subjects in the intervention group received autologous platelet-derived mitochondria through the intracoronary injection. We observed that within 40 days, the intervention group had a slightly greater improvement in the left ventricular ejection fraction (LVEF) compared to the control group and experienced a significant enhancement in the exercise capacity (p < 0.001). Moreover, major adverse cardiac events (MACE), arrhythmia, fever, and tachycardia were compared between the groups and lack of significant difference marks the safety of mitochondrial transplantation (p > 0.05). Furthermore, the two groups were not significantly distinct as regards the average length of stay for a hospitalisation (p > 0.05). CONCLUSION: We suggest platelet-derived mitochondrial transplantation appears as a beneficial and highly promising therapeutic option for patients of ischaemic heart disease (IHD); however, we are aware that further in-depth studies with larger sample sizes along with longer follow-up periods are necessary for validating the clinical implications of our findings.


Subject(s)
Blood Platelets , Myocardial Ischemia , Humans , Male , Female , Middle Aged , Prospective Studies , Treatment Outcome , Myocardial Ischemia/surgery , Myocardial Ischemia/therapy , ST Elevation Myocardial Infarction/surgery , ST Elevation Myocardial Infarction/therapy , Aged , Mitochondria/transplantation
2.
Iran J Med Sci ; 48(3): 268-276, 2023 05.
Article in English | MEDLINE | ID: mdl-37791335

ABSTRACT

Background: Lipocalin-2 (LCN2) deregulation has been reported in several types of cancer and is implicated in the proliferation, migration, angiogenesis, and progression of tumors. However, its aberrant expression has been rarely studied in nasopharyngeal carcinoma (NPC). In the present study, we investigated the expression of LCN2 in NPC patients. Methods: In this descriptive cross-sectional study, 29 NPC and 20 non-cancerous control paraffin pathology blocks were obtained from the seven-year (2011 to 2018) archive of Razi Laboratory in Rasht, Iran. LCN2 mRNA expression was evaluated through quantitative real-time PCR. In addition, immunohistochemistry was performed to evaluate LCN2 expression at the protein level. The fold change value and total immunostaining score (TIS) were applied for quantitative evaluation. The nonparametric Mann-Whitney U test and Fisher's exact test were used through GraphPad Prism 8.3.0 software. P<0.05 was considered statistically significant. Results: Our results revealed that LCN2 mRNA and protein levels in NPC tissues were significantly higher than control tissues (P=0.028 and P=0.002, respectively). At the protein level, 65.51% (19/29) of NPC patients were categorized as having high LCN2 expression (TIS>3) and 34.47% (10/29) as low expression (TIS≤3). While in the control group, 25% (5/20) of subjects represented a high expression of LCN2 (TIS>3), and 75% (15/20) showed no or weak expression (TIS≤3). No significant correlation was found between the overexpression of LCN2 at the protein level and the demographic features of the patients. Conclusion: Our findings suggest that LCN2 might be considered a potential new diagnostic marker for NPC. However, this warrants further studies.


Subject(s)
Nasopharyngeal Neoplasms , Humans , Nasopharyngeal Carcinoma/diagnosis , Nasopharyngeal Carcinoma/genetics , Lipocalin-2/genetics , Lipocalin-2/metabolism , Up-Regulation , Cross-Sectional Studies , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , RNA, Messenger/metabolism , Biomarkers
3.
Int Immunopharmacol ; 118: 110106, 2023 May.
Article in English | MEDLINE | ID: mdl-37015158

ABSTRACT

Each year, traumatic brain injury (TBI) causes a high rate of mortality throughout the world and those who survive have lasting disabilities. Given that the brain is a particularly dynamic organ with a high energy consumption rate, the inefficiency of current TBI treatment options highlights the necessity of repairing damaged brain tissue at the cellular and molecular levels, which according to research is aggravated due to ATP deficiency and reactive oxygen species surplus. Taking into account that mitochondria contribute to generating energy and controlling cellular stress, mitochondrial transplantation as a new treatment approach has lately reduced complications in a number of diseases by supplying healthy and functional mitochondria to the damaged tissue. For this reason, in this study, we used this technique to transplant human umbilical cord-derived mesenchymal stem cells (hUC-MSCs)-derived mitochondria as a suitable source for mitochondrial isolation into rat models of TBI to examine its therapeutic benefit and the results showed that the successful mitochondrial internalisation in the neuronal cells significantly reduced the number of brain cells undergoing apoptosis, alleviated astrogliosis and microglia activation, retained normal brain morphology and cytoarchitecture, and improved sensorimotor functions in a rat model of TBI. These data indicate that human umbilical cord-derived mesenchymal stem cells-isolated mitochondrial transplantation improves motor function in a rat model of TBI via rescuing neuronal cells from apoptosis and alleviating astrogliosis and microglia activation, maybe as a result of restoring the lost mitochondrial content.


Subject(s)
Brain Injuries, Traumatic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Rats , Animals , Gliosis , Microglia , Mitochondria , Apoptosis/physiology , Umbilical Cord
4.
Hum Cell ; 36(3): 963-971, 2023 May.
Article in English | MEDLINE | ID: mdl-36745313

ABSTRACT

To understand the molecular mechanisms responsible for radioresistance in cancer cells, we previously established clinically relevant radioresistant (CRR) cell lines from several human cancer cell lines. These CRR cells proliferate even under exposure to 2 Gy/day of X-rays for more than 30 days, which is a standard protocol for tumor radiotherapy. CRR cells received 2 Gy/day of X-rays to maintain their radioresistance (maintenance irradiation; MI). Interestingly, CRR cells that did not receive MI for more than a year lost their radioresistance, indicating that radiation-induced radioresistance is reversible. We designated these CRR-NoIR cells. Karyotyping of the parental and CRR cells revealed that the chromosomal composition of CRR cells is quite different from that of the parental cells. However, CRR and CRR-NoIR cells were more similar compared with the parental cells because CRR cells repair X-ray-induced DNA damage with higher fidelity. To identify the factor(s) involved in tumor radioresistance, previously published studies including ours have compared radioresistant cells to parental cells. In this review, we conclude that a comparison between CRR and CRR-NoIR cells, rather than parental cells, is the best way to identify factors involved in tumor radioresistance.


Subject(s)
Neoplasms , Radiation Tolerance , Humans , Cell Line, Tumor , Radiation Tolerance/genetics , X-Rays , DNA Damage , Neoplasms/genetics , Neoplasms/radiotherapy
5.
Discov Oncol ; 13(1): 113, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36305981

ABSTRACT

Glioblastoma (GBM) is one of the most difficult cancers to treat because GBM has the high therapeutic resistance. Recently, immunotherapies for GBM have been used instead of conventional treatments. Among them, Natural killer (NK) cell-based immunotherapy has the potential to treat GBM due to its properties such as the absence of restriction by antigen-antibody reaction and deep penetration into the tumor microenvironment. Especially, genetically engineered NK cells, such as chimeric antigen receptor (CAR)-NK cells, dual antigen-targeting CAR NK cells, and adapter chimeric antigen receptor NK cells are considered to be an important tool for GBM immunotherapy. Therefore, this review describes the recent efforts of NK cell-based immunotherapy in GBM patients. We also describe key receptors expressing on NK cells such as killer cell immunoglobulin-like receptor, CD16, and natural killer group 2, member D (NKG2DL) receptor and discuss the function and importance of these molecules.

6.
Adv Pharm Bull ; 12(3): 550-560, 2022 May.
Article in English | MEDLINE | ID: mdl-35935055

ABSTRACT

Purpose: Currently, several disorders including burns, trauma, excisional and diabetic wounds, and bedsores threaten the human health. Application of mesenchymal stem cells (MSCs) is recommended for treatment of skin disorders. However, because of oxidative stress and inflammation after skin injury, survival of transplanted MSCs is low which in turn negatively affects the efficiency of the MSCs-based therapy. In an attempt to address the aforementioned challenge and introducing a novel potential therapeutic strategy, we employed combination therapy by lipocalin 2 (Lcn2)-engineered MSCs and a Metadichol (an inverse agonist of vitamin D receptor (VDR)) nanogel in a rat model of excisional wound. Methods: First, human umbilical cord MSCs (hUC-MSCs) was transfected by a recombinant plasmid encoding Lcn2 gene. Next, a combination of Metadichol nanogel and the engineered MSCs was co-applied on wound in rat model of excision injury. Finally the improvement of wound healing in experimental groups was evaluated by photography and histological assessments (hematoxylin and eosin staining). Results: Our findings revealed that the repair rate was higher in the group received combination therapy comparing to control groups. Notably, Metadichol+Lcn2-MSCs showed significantly higher wound contraction rate compared to control group at all time points (P value < 0.001). Furthermore, wound repair rate was 95% 14 days after surgery, and 100% after 21 days in the treatment groups. Our results also revealed that the combination therapy improved and accelerated the wound healing process. Conclusion: Our findings suggest a novel potential therapeutic strategy i.e. Lcn2-engineered MSCs and Metadichol for wound healing. However, further preclinical and clinical studies are required.

7.
Med Oncol ; 39(11): 175, 2022 Aug 16.
Article in English | MEDLINE | ID: mdl-35972707

ABSTRACT

Breast cancer is the most common type of neoplasm and the second cause of cancer-related death in women. Despite the development of novel therapeutic strategies and improved the clinical outcomes, the mortality rate for breast cancer is still high. Therefore, development of a new modality, particularly based on knocking out key genes, is under focus of investigation. Heme oxygenase-1 (HO-1) deregulation has been associated with various neoplasms-related behaviors of many types of tumor cells including breast cancer. In the current study, in order to evaluate the role of the HO-1 gene in breast cancer, we utilized the CRISPR/Cas9 technology to knock out HO-1 gene in T47D breast cancer cell line and studied its potential therapeutic effects in vitro. The cell proliferation and their sensitivity to Cisplatin were determined by CCK-8 kit. In addition, the apoptosis and the migratory potential of the cells were evaluated using Hoechst staining, and Transwell/Scratch methods, respectively. Our findings revealed that HO-1 suppression significantly reduced the proliferation ability of T47D cells (P < 0.001). Moreover, sensitivity to Cisplatin-induced toxicity increased significantly in KO-T47D cells compared to the control T47D cells. Furthermore, our findings indicated that Cisplatin-induced apoptosis increased in the KO-T47D cells. Moreover, the migratory capability of KO-T47D cells was abolished significantly (P < 0.001) as determined by Transwell migration assay. In a nutshell, our findings strongly suggest that HO-1 involved in breast cancer progression and metastasis and chemotherapy resistance. However, further comprehensive studies are required to clarify the precise role of the HO-1 gene on breast cancer cells.


Subject(s)
Breast Neoplasms , Cisplatin , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Cisplatin/therapeutic use , Female , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans
8.
J Tissue Viability ; 30(4): 527-536, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34366213

ABSTRACT

AIM OF THE STUDY: Skin wounds are a major public health issue due to the lack of real effective remedies. Mesenchymal stem cells (MSCs) are considered as a promising therapeutic strategy for wound injuries; however, low survival rate following transplantation limited their application. In an attempt to introduce a novel potential wound dressing and improve wound healing properties, the current study was conducted. MATERIAL AND METHODS: we prepared conditioned medium (CM) harvested from HEK-293 cells overexpressing nuclear factor erythroid 2-related factor 2 (NRF2), a master regulator of antioxidant genes expression. Then, the CM was loaded in a biodegradable hydrogel. Next, in an animal model of full-thickness excision wound, wharton's jelly derived-mesenchymal stem cells (WJ-MSCs) were transplanted at the margins of the wound followed by application of the hydrogel on injury site. Finally, wound healing characteristics were evaluated by proper methods. RESULTS: Our findings revealed that, the NRF2-CM protected the WJ-MSCs against H2O2-induced toxicity in vitro. Furthermore, in vivo results showed that, SA/G hydrogel containing NRF2-CM significantly (P < 0.01) promoted WJ-MSCs survival, increased angiogenesis, accelerated wound contraction, and promoted wound healing compared to other groups. CONCLUSION: Though further preclinical and clinical studies regarding mechanisms behind the protection and also safety of the strategy are needed, our findings strongly suggest that the prepared wound dressing enhanced the efficacy of therapeutic potential of WJ-MSCs by providing an enriched/antioxidant niche support.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Culture Media, Conditioned , HEK293 Cells , Humans , Hydrogels , Hydrogen Peroxide , NF-E2-Related Factor 2 , Rats , Wound Healing
10.
Appl Biochem Biotechnol ; 191(3): 1056-1071, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31956957

ABSTRACT

Cartilage hypertrophy is a condition in which the cells are completely differentiated, and new morphological changes and mineralization prevent proper cellular functions. The occurrence of hypertrophy during differentiation fails current regenerative strategies for treatment. Strategies to minimize hypertrophy in chondrocytes are categorized into two levels of protein and gene. Among these strategies, one way to affect multiple pathways involved in the development of hypertrophy is to manage microRNA activity in cells. Recent miRNA profiling studies have shown that miR-195-5p upregulates through the transition from chondrogenic to hypertrophic state. Bioinformatics assessment of microRNA targets also indicates that several genes repressed by miR-195-5p play important roles in processes related to hypertrophy. The aim of this study was to develop a microRNA Tough Decoy to suppress miR-195-5p and investigate whether it can prevent a hypertrophic state in chondrocytes. The Tough Decoy (TUD) was designed and evaluated bioinformatically and then cloned into the pLVX-Puro plasmid. The TUD function was validated by Dual-Luciferase assay and qRT-PCR. After delivering TUD to C28/I2 chondrocytes cultured in a hypertrophic medium, hypertrophic differentiation was assessed by histochemical staining, quantitative RT-PCR of hypertrophy marker genes, and alkaline phosphatase activity. Results showed that the TUD could inhibit miRNA efficiently and downregulate hypertrophic markers such as RUNX2, alkaline phosphatase, and collagen 10 significantly compared with the control group. Alcian blue and alizarin red staining also demonstrated the optimal effect of gene constructs on tissue properties and mineralization of the TUD group. Delivering the miR-195-5p Tough Decoy to the cartilage cells can prevent the occurrence of hypertrophy in chondrocytes and could be considered as a candidate for the treatment of other diseases such as osteoarthritis.


Subject(s)
Chondrocytes/cytology , MicroRNAs/genetics , Osteoarthritis/therapy , Alkaline Phosphatase/metabolism , Cell Survival , Cells, Cultured , Computational Biology , Humans , Hypertrophy/metabolism , Osteoarthritis/metabolism , Phenotype , Plasmids/genetics , Regeneration , Signal Transduction , Up-Regulation
11.
Iran J Med Sci ; 44(6): 493-500, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31875084

ABSTRACT

BACKGROUND: Due to oxidative stress, hypoxia, and serum deprivation, a large percentage of mesenchymal stem cells (MSCs) die in the early stages of transplantation. The present study aimed to address whether induction or inhibition of autophagy would affect the viability of MSCs after exposure to oxidative stress. METHODS: MSCs were isolated from umbilical cord tissue using the Ficoll gradient method. pCMV-GFP-LC-3 plasmid containing GFP-tagged LC3 was transfected into MSCs to assay autophagy level in these valuable cells. The four study groups were: MSC-LC3-Rapa, MSC-LC3-3MA, MSCs without any transfection, and MSC-GFP-LC3 (control groups). To induce autophagy, the MSC-GFP-LC3 was treated with different concentrations of Rapa for 24 hours and named MSC-LC3-Rapa. To inhibit autophagy in MSC-GFP-LC3, these cells were cultured in the presence of 3MA for 24 hours and named MSC-LC3-3MA. Non-treated MSC-GFP-LC3 and MSCs were considered as control groups. MSCs were exposed to lethal doses of H2O2 followed by cell viability evaluation with the water-soluble tetrazolium salt assay method. The data were analyzed with SPSS version 18.0 using one-way ANOVA test. P<0.05 was considered statistically significant. RESULTS: The results revealed that the enhancement of autophagy in MSC-LC3-Rapa sensitized them against oxidative stress (P=0.0006) and inhibition of autophagy in MSC-LC3-3MA led to resistance against oxidative stress (P=0.0003). CONCLUSION: Inhibition of autophagy, as a non-genetic engineering method, in MSCs enhances cell viability following exposure to the oxidative stress. This may provide a novel strategy to promote the efficiency of MSC-based cell therapy for clinical applications.

12.
Mol Biol Rep ; 46(1): 1533-1549, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30623280

ABSTRACT

In autoimmune disease body's own immune system knows healthy cells as undesired and foreign cells. Over 80 types of autoimmune diseases have been recognized. Currently, at clinical practice, treatment strategies for autoimmune disorders are based on relieving symptoms and preventing difficulties. In other words, there is no effective and useful therapy up to now. It has been well-known that mesenchymal stem cells (MSCs) possess immunomodulatory effects. This strongly suggests that MSCs might be as a novel modality for treatment of autoimmune diseases. Supporting this notion a few preclinical and clinical studies indicate that MSCs ameliorate autoimmune disorders. Interestingly, it has been found that the beneficial effects of MSCs in autoimmune disorders are not relying only on direct cell-to-cell communication but on their capability to produce a broad range of paracrine factors including growth factors, cytokines and extracellular vehicles (EVs). EVs are multi-signal messengers that play a serious role in intercellular signaling through carrying cargo such as mRNA, miRNA, and proteins. Numerous studies have shown that MSC-derived EVs are able to mimic the effects of the cell of origin on immune cells. In this review, we discuss the current studies dealing with MSC-based therapies in autoimmune diseases and provide a vision and highlight in order to introduce MSC-derived EVs as an alternative and emerging modality for autoimmune disorders.


Subject(s)
Autoimmune Diseases/therapy , Extracellular Vesicles/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Clinical Trials as Topic , Humans , Immunosuppression Therapy
13.
Adv Pharm Bull ; 8(3): 465-470, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30276143

ABSTRACT

Purpose: Poor survival rate of mesenchymal stem cells (MSCs) following their transplantation is one of the major challenges in their therapeutic application. Therefore, it is necessary to augment the viability of the MSCs in order to improve their therapeutic efficacy. Several strategies have been used to overcome this problem. Preconditioning of MSCs with oxidative stresses has gained a lot of attention. Therefore, in the present study, we investigated the effects of simultaneous preconditioning of MSCs with hydrogen peroxide and serum deprivation stresses on their survival and resistance to stressful conditions. Methods: MSCs were isolated from human umbilical cord blood. To perform simultaneous preconditioning, the cells were cultured in DMEM medium containing 1, 2.5 and 5 percent FBS and different concentrations of H2O2 (5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 80 and 100 µM) for 24 hrs. Then, the cells were cultured in recovery culture medium. Finally, one group of the cells was exposed to a lethal concentration of H2O2 (300µM), and the other cells were cultivated in FBS free DMEM medium as the lethal situation. In addition, the percentage of apoptotic cells was analyzed using Caspase 3 assay kit. Results: Simultaneous preconditioning of the MSCs with 15µM H2O2 plus serum deprivation, 2.5% FBS, significantly increased the resistance of the cells to the toxicity induced following their cultivation in FBS free DMEM medium. It exerted the protective effect on the cells after treating with the lethal dose of H2O2 as well. Conclusion: Simultaneous preconditioning of MSCs with oxidative and serum deprivation stresses enhances their survival against harsh conditions, which might increase the viability and stability of the MSCs following their transplantation.

14.
EXCLI J ; 17: 590-597, 2018.
Article in English | MEDLINE | ID: mdl-30108463

ABSTRACT

The NGR peptide is one of the well-known peptides for targeting tumor cells. It has the ability to target aminopeptidase N (CD13) on tumor cells or the tumor vascular endothelium. In this study, the NGR peptide was used for targeting A subunit of the Shiga toxin to cancer cells. The cytotoxic effect of the A-NGR fusion protein was assessed on HT1080, U937, HT29 cancer cells and MRC-5 normal cells. For this purpose, cells were treated with different concentrations of A-NGR (0.5-40 µg/ml). The evaluation of cell viability was achieved by MTT assay. Apoptosis was determined by annexin-V/PI double staining flow cytometry. Alterations in the mRNA expression of apoptosis - related genes were assessed by real time RT- PCR. The results showed that A-NGR fusion protein effectively inhibited the growth of HT1080 and U937 cancer cells in comparison to negative control (PBS) but for CD13-negative HT-29 cancer cells, only at high concentrations of fusion protein was inhibited growth recorded. On the other hand, A-NGR had little cytotoxic effect on MRC-5 normal cells. The flow cytometry results showed that A-NGR induces apoptosis. Furthermore, the results of real time RT-PCR revealed that A-NGR significantly increases the mRNA expression of caspase 3 and caspase 9. Conclusively, A-NGR fusion protein has the ability of targeting CD13-positive cancer cells, the cytotoxic effect on CD13-positive cancer cells as well as has low cytotoxic effect on normal cells.

15.
Iran J Pharm Res ; 17(1): 147-154, 2018.
Article in English | MEDLINE | ID: mdl-29755547

ABSTRACT

Cisplatin is one of the most useful chemotherapeutics which performs its cytotoxic effect via accumulation of platinum resulting in oxidative stress, and destruction of cell DNA. This could probably cause secondary cancers in healthy tissues. Lipocalin2 (Lcn2) is a protein which its expression is increased in oxidative stresses. Therefore, the present study was performed to evaluate the protective effects of Lcn2 up-regulation on cisplatin genotoxicity. In order to up-regulate Lcn2 expression, HEK293 cells were transfected with pcDNA3.1-Lcn2 vector. Afterwards, stable cells consistently expressing Lcn2 were selected via screening with G418 antibiotic. Next, overexpression of Lcn2 was evaluated by RT-PCR and ELISA, comparing to the control non-transfected cells. Then, in order to evaluate the cytoprotective effects of Lcn2 overexpression, transfected and non-transfected cells were subjected to cisplatin treatment followed by MTT and alkaline Comet assays. RT-PCR and ELISA assays confirmed up-regulation of Lcn2 by the stable cells. MTT assay of the Lcn2 over-expressing cells showed higher IC50 values comparing to the non-transfected cells. Furthermore, the Comet assay confirmed Lcn2 protective effects on the cisplatin (1 µg/mL) induced genotoxicity. In the present study, for the first time, we showed the protective effect of Lcn2 on cisplatin induced genotoxicity. Therefore, one of the probable mechanisms of Lcn2 cytoprotctive effects under oxidative stress conditions could be due to the prevention of genotoxicity. However, further evaluations in this regard must be considered.

16.
Artif Organs ; 40(12): E266-E279, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27911032

ABSTRACT

Transforming growth factor beta-3 (TGF-ß3) has been shown to decrease scar formation after scheduled topical applications to the cutaneous wounds. This study aimed to continuously deliver TGF-ß3, during the early phase of wound healing, by engineering a dermal equivalent (DE) using TGF-ß3 expressing bone marrow stromal cells (BM-SCs) and human dehydrated amniotic membrane (hDAM). To engineer a DE, rat BM-SCs were seeded on the hDAM and TGF-ß3 was transiently transfected into the BM-SCs using a plasmid vector. Pieces of the dermal equivalent were transplanted onto the full-thickness excisional skin wounds in rats. The process of wound healing was assessed by image analysis, Manchester Scar Scale (MSS), and histopathological studies 7, 14, 21, and 85 days after the excision. The results confirmed accurate construction of recombinant pcDNA3.1-TGF-ß3 expression system and showed that the transfected BM-SCs seeded on hDAM expressed TGF-ß3 mRNA and protein from day 3 through day 7 after transfection. After implantation of the DE, contraction of the wounds was measured from day 7 through 21 and analyzed by linear regression, which revealed that the rate of wound contraction in all experimental groups was similar. Histologic evaluation demonstrated that transfected BM-SCs decreased retention and recruitment of the cells during the early stage of wound healing, decreased the formation of vascular structures and led to formation of uniformly parallel collagen bundles. MSS scores showed that TGF-ß3 secreting cells significantly improved the cosmetic appearance of the healed skin and decreased the scar formation. From these results, it could be concluded that transient secretion of TGF-ß3, during the early phase of healing, by BM-SCs seeded on hDAM can improve the cosmetic appearance of the scar in cutaneous wounds without negatively affecting the process of wound repair.


Subject(s)
Amnion/chemistry , Mesenchymal Stem Cells/cytology , Skin/pathology , Tissue Scaffolds/chemistry , Transforming Growth Factor beta3/genetics , Wound Healing , Amnion/cytology , Animals , Bioprosthesis , Cells, Cultured , Female , Gene Expression , Genetic Engineering , Genetic Vectors/genetics , Humans , Mesenchymal Stem Cells/metabolism , Plasmids/genetics , Rats , Rats, Wistar , Skin/injuries , Skin/ultrastructure , Skin, Artificial , Transfection
17.
Iran Biomed J ; 20(3): 135-44, 2016 07.
Article in English | MEDLINE | ID: mdl-26899739

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have been recently received increasing attention for cell-based therapy, especially in regenerative medicine. However, the low survival rate of these cells restricts their therapeutic applications. It is hypothesized that autophagy might play an important role in cellular homeostasis and survival. This study aims to investigate the regenerative potentials of autophagy-modulated MSCs for the treatment of acute liver failure (ALF) in mice. METHODS: ALF was induced in mice by intraperitoneal injection of 1.5 ml/kg carbon tetrachloride. Mice were intravenously infused with MSCs, which were suppressed in their autophagy pathway. Blood and liver samples were collected at different intervals (24, 48 and 72 h) after the transplantation of MSCs. Both the liver enzymes and tissue necrosis levels were evaluated using biochemical and histopathological assessments. The survival rate of the transplanted mice was also recorded during one week. RESULTS: Biochemical and pathological results indicated that 1.5 ml/kg carbon tetrachloride induces ALF in mice. A significant reduction of liver enzymes and necrosis score were observed in autophagy-modulated MSC-transplanted mice compared to sham (with no cell therapy) after 24 h. After 72 h, liver enzymes reached their normal levels in mice transplanted with autophagy-suppressed MSCs. Interestingly, normal histology without necrosis was also observed. CONCLUSION: Autophagy suppression in MSCs ameliorates their liver regeneration potentials due to paracrine effects and might be suggested as a new strategy for the improvement of cell therapy in ALF.


Subject(s)
Autophagy/physiology , Cell- and Tissue-Based Therapy/methods , Liver Failure, Acute/therapy , Liver Regeneration/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , Autophagy-Related Protein 7/metabolism , Bone Marrow Cells/metabolism , Carbon Tetrachloride/toxicity , Cell Differentiation , Cell Proliferation , Cell Survival , Disease Models, Animal , Humans , Liver/enzymology , Liver/pathology , Liver Failure, Acute/pathology , Mice
18.
Vaccine ; 34(12): 1472-9, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26868080

ABSTRACT

Flagellin of Pseudomonas aeruginosa is an important vaccine candidate. N-terminal domains are highly conserved in both type a and type b flagellins. The efficacy of gold nanoparticles (AuNPs) conjugated to N-terminal domains of P. aeruginosa flagellin (flagellin(1-161)), as an immunogen in mice, has been assessed. The nanoparticles were conjugated to the recombinant protein through direct interaction of thiol molecules of the cysteines with AuNPs and formation of AuS bond. Flagellin(1-161), AuNP-flagellin(1-161), and flagellin(1-161) emulsified in Freund's adjuvant (FA: complete/incomplete Freund's adjuvant formulation) were administered subcutaneously to BALB/c mice. Mice given AuNP-flagellin(1-161) elicited high titers of anti-flagellin(1-161) antibodies compared with non-immune group and/or mice which received flagellin(1-161) without adjuvant. In whole cell ELISA, these antibodies effectively recognized the native flagellin on the bacteria. Opsonophagocytosis assay demonstrated the functional activity and specificity of anti-flagellin(1-161) antibodies raised by AuNP-flagellin(1-161) against homologous strain. All of the results were comparable with those obtained by use of FA. Taken together, this is the first report of conjugation of AuNPs to flagellin and evaluating its immune response against P. aeruginosa.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Flagellin/immunology , Immunity, Humoral , Metal Nanoparticles/administration & dosage , Animals , Antibodies, Bacterial/blood , Cells, Cultured , Female , Freund's Adjuvant/administration & dosage , Gold , Lipids/administration & dosage , Macrophages/immunology , Mice, Inbred BALB C , Phagocytosis , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa , Recombinant Proteins/immunology
19.
Adv Pharm Bull ; 5(2): 201-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26236658

ABSTRACT

PURPOSE: Recent developments in the field of cell therapy have led to a renewed interest in treatment of acute kidney injury (AKI). However, the early death of transplanted mesenchymal stem cells (MSCs) in stressful microenvironment of a recipient tissue is a major problem with this kind of treatment. The objective of this study was to determine whether overexpression of a cytoprotective factor, nuclear factor erythroid-2 related factor 2 (Nrf2), in MSCs could protect rats against AKI. METHODS: The Nrf2 was overexpressed in MSCs by recombinant adenoviruses, and the MSCs were implanted to rats suffering from cisplatin-induced AKI. RESULTS: The obtained results showed that transplantation with the engineered MSCs ameliorates cisplatin-induced AKI. Morphologic features of the investigated kidneys showed that transplantation with the MSCs in which Nrf2 had been overexpressed significantly improved the complications of AKI. CONCLUSION: These findings suggested that the engineered MSCs might be a good candidate to be further evaluated in clinical trials. However, detailed studies must be performed to investigate the possible carcinogenic effect of Nrf2 overexpression.

20.
Iran J Basic Med Sci ; 18(5): 459-64, 2015 May.
Article in English | MEDLINE | ID: mdl-26124931

ABSTRACT

OBJECTIVES: Lipocalin2 (Lcn2) gene is highly expressed in response to various types of cellular stresses. The precise role of Lcn2 has not been fully understood yet. However, it plays a key role in controlling vital cellular processes such as proliferation, apoptosis and metabolism. Recently it was shown that Lcn2 decreases senescence and increases proliferation of mesenchymal stem cells (MSC) with finite life span under either normal or oxidative stress conditions. However, Lcn2 effects on immortal cell line with infinite proliferation are not defined completely. Materials and. MATERIALS AND METHODS: HEK-293 cells were transfected with recombinant pcDNA3.1 containing Lcn2 fragment (pcDNA3.1-Lcn2). Expression of lipocalin2 in transfected cells was evaluated by RT-PCR, real time RT-PCR, and ELISA. Different cell groups were treated with H2O2 and WST-1 assay was performed to determine their proliferation rate. Senescence was studied by ß-galactosidase and gimsa staining methods as well as evaluation of the expression of senescence-related genes by real time RT-PCR. RESULTS: Lcn2 increased cell proliferation under normal culture condition, while the proliferation slightly decreased under oxidative stress. This decrease was further found to be attributed to senescence. CONCLUSION: Our findings indicated that under harmful conditions, Lcn2 gene is responsible for the regulation of cell survival through senescence.

SELECTION OF CITATIONS
SEARCH DETAIL
...