Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Stem Cell Res Ther ; 13(1): 217, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35619143

ABSTRACT

Retinal pigment epithelium (RPE) degeneration is the hallmark of age-related macular degeneration (AMD). AMD, as one of the most common causes of irreversible visual impairment worldwide, remains in need of an appropriate approach to restore retinal function. Wet AMD, which is characterized by neovascular formation, can be stabilized by currently available therapies, including laser photocoagulation, photodynamic therapy, and intraocular injections of anti-VEFG (anti-vascular endothelial growth factor) therapy or a combination of these modalities. Unlike wet AMD, there is no effective therapy for progressive dry (non-neovascular) AMD. However, stem cell-based therapies, a part of regenerative medicine, have shown promising results for retinal degenerative diseases such as AMD. The goal of RPE cell therapy is to return the normal structure and function of the retina by re-establishing its interaction with photoreceptors, which is essential to vision. Considering the limited source of naturally occurring RPE cells, recent progress in stem cell research has allowed the generation of RPE cells from human pluripotent cells, both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSC). Since iPSCs face neither ethical arguments nor significant immunological considerations when compared to ESCs, they open a new horizon for cell therapy of AMD. The current study aims to discuss AMD, review the protocols for making human iPSCs-derived RPEs, and summarize recent developments in the field of iPSC-derived RPEs cell therapy.


Subject(s)
Induced Pluripotent Stem Cells , Macular Degeneration , Cell- and Tissue-Based Therapy , Epithelium/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Macular Degeneration/metabolism , Macular Degeneration/therapy , Retinal Pigment Epithelium/metabolism
2.
Pediatr Blood Cancer ; 69(8): e29676, 2022 08.
Article in English | MEDLINE | ID: mdl-35441789

ABSTRACT

BACKGROUND: Natural killer (NK) cell therapy has been shown to be effective in the treatment of some cancers. However, the effects of this adoptive immunotherapy have not been investigated for Wilms tumor (WT). In this study, the effects of adoptive NK-cell transfer on a patient-derived xenograft (PDX) model of anaplastic WT were evaluated, and the impacts of cell source and ex vivo activation strategy on the therapeutic efficacy of NK-cell product were appraised. METHODS: NK cells were isolated from human peripheral blood mononuclear cells (NKPB ) and human cord blood (NKCB ), and were expanded and activated using a cytokine cocktail. Another group of NK cells (NKET ) was produced through activation with the exosomes extracted from previously challenged NKPB cells with WT. PDX-bearing mice were treated with clinically relevant doses of NKPB , NKCB , NKET , standard chemotherapy, and placebo (phosphate-buffered saline). RESULTS: PDX models treated with NKCB showed a better survival rate, though the difference among the study groups was not significant. Compared with the placebo control group, NKCB significantly improved the histopathologic response, NKPB significantly inhibited the proliferation of neoplastic cells, and NKET led to a significant decrease in the metastasis score (all p-values <.05). Standard chemotherapy provided the greatest tumor growth inhibition and the lowest mitotic count, though it did not show any significant advantage over NK-cell therapies in any of the outcome parameters in two-by-two comparisons. CONCLUSIONS: This study spotlights the efficacy of adoptive NK-cell transfer as a potential treatment candidate for high-risk WT.


Subject(s)
Kidney Neoplasms , Wilms Tumor , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Immunotherapy, Adoptive , Kidney Neoplasms/therapy , Killer Cells, Natural/transplantation , Leukocytes, Mononuclear , Mice , Wilms Tumor/therapy
3.
Neurol Sci ; 43(1): 399-410, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34032944

ABSTRACT

INTRODUCTION: Spinal muscular atrophy (SMA), an autosomal recessive neurodegenerative disorder of alpha motor neurons of spinal cord associated with progressive muscle weakness and hypotonia, is the most common genetic cause of infant mortality. Although there is few promising treatment for SMA, but the field of translational research is active in it, and stem cell-based therapy clinical trials or case studies are ongoing. Combination of different therapeutic approaches for noncurative treatments may increase their effectiveness and compliance of patients. We present a phase 1 clinical trial in patients with SMA1 who received side population adipose-derived mesenchymal stem cells (SPADMSCs). METHODS: The intervention group received three intrathecal administrations of escalating doses of SPADMSCs and followed until 24 months or the survival time. The safety analysis was assessed by controlling the side effects and efficacy evaluations performed by the Hammersmith Infant Neurological Examination (HINE), Ballard score, and electrodiagnostic (EDX) evaluation. These evaluations were performed before intervention and at the end of the follow-up. RESULTS: The treatment was safe and well tolerated, without any adverse event related to the stem cell administration. One of the patients in the intervention group was alive after 24 months of study follow-up. He is a non-sitter 62-month-old boy with appropriate weight gain and need for noninvasive ventilation (NIV) for about 8 h per day. Clinical scores, need for supportive ventilation, and number of hospitalizations were not meaningful parameters in the response of patients in the intervention and control groups. All five patients in the intervention group showed significant improvement in the motor amplitude response of the tibial nerve (0.56mV; p: 0.029). CONCLUSION: This study showed that SPADMSCs therapy is tolerable and safe with promising efficacy in SMA I. Probably same as other treatment strategies, early intervention will increase its efficacy and prepare time for more injections. We suggest EDX evaluation for the follow-up of treatment efficacy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cells , Muscular Atrophy, Spinal , Spinal Muscular Atrophies of Childhood , Child, Preschool , Humans , Male , Spinal Muscular Atrophies of Childhood/therapy , Treatment Outcome
4.
Biomed Res Int ; 2021: 5597758, 2021.
Article in English | MEDLINE | ID: mdl-34124249

ABSTRACT

BACKGROUND: Investigating the viability and proliferative rates of fibroblast cells on human amniotic membrane (HAM) as a scaffold will be an important subject for further research. The aim of this study was to assess the fibroblast viability seeded on acellular HAM, since foreskin neonatal allogenic fibroblasts seeded on HAM accelerate the wound healing process. METHODS: Fibroblasts were retrieved from the foreskin of a genetically healthy male infant, and we exploited AM of healthy term neonates to prepare the amniotic scaffold for fibroblast transfer. After cell culture, preparation of acellular HAM, and seeding of cells on HAM based on the protocol, different methods including 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 4',6-Diamidino-2-phenylindole dihydrochloride (DAPI), and propidium iodide (PI) staining were employed for assessment of fibroblast viability on HAM. RESULTS: Based on the results obtained from the DAPI and PI staining, the percentage of viable cells in the former staining was clearly higher than that of the dead cells in the latter one. The results of DAPI and PI staining were in accordance with the findings of MTT assay, confirming that fibroblasts were viable and even proliferate on HAM. CONCLUSION: Our findings showed the viability of fibroblasts seeded on the acellular HAM using MTT assay, DAPI, and PI staining; however, this study had some limitations. It would be an interesting subject for future research to compare the viability and proliferation rate of fibroblasts seeded on both cellular and acellular HAM.


Subject(s)
Amnion/chemistry , Cell Culture Techniques , Fibroblasts/metabolism , Tissue Scaffolds/chemistry , Cell Survival , Fibroblasts/cytology , Humans , Male
5.
Reprod Biol ; 21(2): 100472, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33639342

ABSTRACT

Female reproductive system disorders (FRSD) with or without infertility are prevalent women's health problems with a variety of treatment approaches including surgery and hormone therapy. It currently considering to sub-branch of regenerative medicine including stem cells or growth factors injection-based delivery treatment might be improved female reproductive health life. The most common products used for these patients treatment are autologous cell or platelet-based products from patients, including platelet-rich plasma, plasma rich in growth factor, platelet-rich fibrin, and stromal vascular fraction. In this review, we discuss each of the above products used in treatment of FRSD and critically evaluate the clinical outcome.


Subject(s)
Infertility, Female/therapy , Stem Cell Transplantation , Stem Cells/classification , Female , Humans , Regenerative Medicine , Stem Cells/physiology
6.
J Mol Neurosci ; 67(2): 247-257, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30535775

ABSTRACT

Cell therapy and stem cell transplantation strategies have provided potential therapeutic approaches for the treatment of neurological disorders. Adipose-derived mesenchymal stem cells (ADMSCs) are abundant adult stem cells with low immunogenicity, which can be used for allogeneic cell replacement therapies. Differentiation of ADMSCs into acetylcholine-secreting motoneurons (MNs) is a promising treatment for MN diseases, such as spinal muscular atrophy (SMA), which is associated with the level of SMN1 gene expression. The SMN2 gene plays an important role in MN disorders, as it can somewhat compensate for the lack of SMN1 expression in SMA patients. Although the differentiation potential of ADMSCs into MNs has been previously established, overexpression of SMN2 gene in a shorter period with a longer survival has yet to be elucidated. Ponasterone A (PNA), an ecdysteroid hormone activating the PI3K/Akt pathway, was studied as a new steroid to promote SMN2 overexpression in MNs differentiated from ADMSCs. After induction with retinoic acid, sonic hedgehog, forskolin, and PNA, MN phenotypes were differentiated from ADMSCs, and immunochemical staining, specific for ß-tubulin, neuron-specific enolase, and choline acetyltransferase, was performed. Also, the results of real-time PCR assay indicated nestin, Pax6, Nkx2.2, Hb9, Olig2, and SMN2 expression in the differentiated cells. After 2 weeks of treatment, cultures supplemented with PNA showed a longer survival and a 1.2-fold increase in the expression of SMN2 (an overall 5.6-fold increase; *P ≤ 0.05), as confirmed by the Western blot analysis. The PNA treatment increased the levels of ChAT, Isl1, Hb9, and Nkx2 expression in MN-like cells. Our findings highlight the role of PNA in the upregulation of SMN2 genes from MSC-derived MN-like cells, which may serve as a potential candidate in cellular therapy for SMA patients.


Subject(s)
Adipocytes/metabolism , Ecdysterone/analogs & derivatives , Mesenchymal Stem Cells/metabolism , Motor Neurons/metabolism , Neurogenesis , Adipocytes/cytology , Adipocytes/drug effects , Adolescent , Adult , Aged , Cells, Cultured , Ecdysterone/pharmacology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Middle Aged , Motor Neurons/cytology , Nuclear Proteins , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism , Transcription Factors , Up-Regulation
7.
Front Immunol ; 8: 1486, 2017.
Article in English | MEDLINE | ID: mdl-29163547

ABSTRACT

There are many types of leukocytes reside in subcutaneous adipose tissue (SAT), and among them, natural killer cells (NKs) comprise a major part. We show that the NKs that reside in the SAT (adipose tissue-derived NK cells; ADNKs) of the abdominal region found with phenotypic differences from the NKs circulating in the peripheral blood derived NK cells (PBNKs). In this survey, flow cytometry phenotyping was used to study the differences between the natural cytotoxicity receptor expression on ADNKs and PBNKs of both obese and lean persons. Also, their cytotoxicity and cytokine production patterns were evaluated. The activation experiments on isolated and expanded NKs with IL-2, IL-15, and IL-21 cytokines revealed the main population of the CD56dim within the total ADNKs of obese persons has an under-expression of NKp30 and NKp44 despite the unchanged levels of NKG2D. The data suggest the suppressive condition of the adipose tissue niche on the NKs response against sensitive major histocompatibility complex class I non-expressing neoplastic cells. As the NKs are the first line of the body's defense vs tumor formation, this change may lead to the development of transformed cells into the tumors.

8.
J Immunother ; 40(7): 265-276, 2017 09.
Article in English | MEDLINE | ID: mdl-28622272

ABSTRACT

Immune cell-derived exosomes can increase immunity against tumors. In contrast, tumor-derived exosomes can reduce the immunity and can change the tumor microenvironment to further develop and provide metastasis. These effects take place by an alteration in the innate and adaptive immune cell functions. In this experiment, we studied the natural killer (NK) cells' effectiveness on tumor cells after expansion and thereafter incubated it with exosomes. The exosomes were derived from 2 populations of NK cells: (1) naive NK cells and, (2) NK cells previously exposed to neuroblastoma (NB) cells. Moreover, we have studied the NB-derived exosomes on NK cell function. The molecular load of the characterized exosomes (by means of nanoparticle-tracking analysis, flow cytometry, scanning electron microscopy, and western blot) from NK cells exposed to the NB cell revealed their expression of natural killer cell receptors in addition to CD56, NKG2D, and KIR2DL2 receptors. These exosomes were used to treat NK cells and thereafter administered to NB tumor cells both in vitro and in vivo. Our results showed some kind of NK cells' education by the exosomes. This education from NK cells previously exposed to NB cell-derived exosomes caused efficient and greater cytotoxicity against NB tumors, but NB-derived exosomes act as tumor promoters by providing a tumor supporting niche. Hence, this method of preparing the exosomes has a dramatic effect on activation of anti-NK cells against NB cells.


Subject(s)
Cytokines/metabolism , Exosomes/metabolism , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neuroblastoma/immunology , Neuroblastoma/metabolism , Animals , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Heterografts , Humans , Immunophenotyping , Male , Mice , Neuroblastoma/mortality , Neuroblastoma/pathology , Phenotype , Receptors, Natural Killer Cell/metabolism , Tumor Burden
9.
Bioimpacts ; 6(3): 149-154, 2016.
Article in English | MEDLINE | ID: mdl-27853678

ABSTRACT

Introduction: The rejuvenation characteristics of fat tissue grafting has been established for many years. Recently it has been shown that stromal vascular fraction (SVF) of fat tissue contributes to its rejuvenation properties. As the SVF is a minimal processed cell population (based on FDA guidance), therefore it is a suitable cell therapy for skin rejuvenation. This clinical trial was aimed to evaluate the ultrastructural improvement of aging skin in the facial nasolabial region after transplantation of autologous SVF. Methods: Our study was conducted in 16 patients aged between 38 and 56 years old that were interested in face lifting at first. All of the cases underwent the lipoaspiration procedure from the abdomen for sampling of fat tissue. Quickly, the SVF was harvested from 100 mL of harvested fat tissue and then transplanted at dose of 2.0×107 nucleated cells in each nasolabial fold. The changes in the skin were evaluated using Visioface scanner, skin-scanner DUB, Visioline, and Cutometer with multi probe adopter. Results: By administration of autologous SVF, the elasticity and density of skin were improved significantly. There were no changes in the epidermis density in scanner results, but we noticed a significant increase in the dermis density and also its thickness with enrichment in the vascular bed of the hypodermis. The score of Visioface scanner showed slight changes in wrinkle scores. The endothelial cells and mesenchymal progenitors from the SVF were found to chang the architecture of the skin slightly, but there was not obvious phenotypic changes in the nasolabial grooves. Conclusion: The current clinical trial showed the modification of dermis region and its microvascular bed, but no changes in the density of the epidermis. Our data represent the rejuvenation process of facial skin by improving the dermal architecture.

10.
Bioimpacts ; 6(2): 99-104, 2016.
Article in English | MEDLINE | ID: mdl-27525227

ABSTRACT

INTRODUCTION: Much attention has been paid to the idea of cell therapy using stem cells from different sources of the body. Fat-derived stem cells that are called adipose derived stem cells (ADSCs) from stromal vascular fraction (SVF) are the subject of many studies in several cell therapy clinical trials. Despite production of some GMP-grade enzymes to isolate SVF for clinical trials, there are critical conditions like inconsistency in lot-to-lot enzyme activity, endotoxin residues, other protease activities and cleavage of some cell surface markers which significantly narrow the options. So we decided to develop a new method via sonication cavitation to homogenize fat tissue and disrupt partially adipose cells to obtain SVF and finally ADSCs at a minimum of time and expenses. METHODS: The fat tissue was chopped in a sterile condition by a blender mixer and then sonicated for 2 s before centrifugation. The next steps were performed as the regular methods of SVF harvesting, and then it was characterized using flow cytometry. RESULTS: Analysis of the surface markers of the cells revealed similar sets of surface antigens. The cells showed slightly high expression of CD34, CD73 and CD105. The differentiation capacity of these cells indicates that multipotent properties of the cells are not compromised after sonication. But we had the less osteogenic potential of cells when compared with the enzymatic method. CONCLUSION: The current protocol based on the sonication-mediated cavitation is a rapid, safe and cost-effective method, which is proposed for isolation of SVF and of course ADSCs cultures in a large scale for the clinical trials or therapeutic purposes.

12.
J Tissue Eng Regen Med ; 9(11): 1268-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-23319462

ABSTRACT

Reconstruction of the bladder wall via in vitro differentiated stem cells on an appropriate scaffold could be used in such conditions as cancer and neurogenic urinary bladder. This study aimed to examine the potential of human endometrial stem cells (EnSCs) to form urinary bladder epithelial cells (urothelium) on nanofibrous silk-collagen scaffolds, for construction of the urinary bladder wall. After passage 4, EnSCs were induced by keratinocyte growth factor (KGF) and epidermal growth factor (EGF) and seeded on electrospun collagen-V, silk and silk-collagen nanofibres. Later we tested urothelium-specific genes and proteins (uroplakin-Ia, uroplakin-Ib, uroplakin-II, uroplakin-III and cytokeratin 20) by immunocytochemistry, RT-PCR and western blot analyses. Scanning electron microscopy (SEM) and histology were used to detect cell-matrix interactions. DMEM/F12 supplemented by KGF and EGF induced EnSCs to express urothelial cell-specific genes and proteins. Either collagen, silk or silk-collagen scaffolds promoted cell proliferation. The nanofibrous silk-collagen scaffolds provided a three-dimensional (3D) structure to maximize cell-matrix penetration and increase differentiation of the EnSCs. Human EnSCs seeded on 3D nanofibrous silk-collagen scaffolds and differentiated to urothelial cells provide a suitable source for potential use in bladder wall reconstruction in women.


Subject(s)
Endometrium/cytology , Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Urinary Bladder/pathology , Urothelium/pathology , Adult , Biocompatible Materials/pharmacology , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Collagen/chemistry , Enzyme-Linked Immunosorbent Assay , Epidermal Growth Factor/metabolism , Female , Fibroblast Growth Factor 7/metabolism , Humans , Immunohistochemistry , Keratinocytes/cytology , Microscopy, Electron, Scanning , Nanofibers/chemistry , Phenotype , Silk/chemistry , Urothelium/cytology
13.
Cell Biol Int ; 39(5): 515-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25490878

ABSTRACT

Today, there is a need for a platform to efficiently generate and maintain a feeder free culture of pluripotent stem cells by small molecules or pharmacological agents. Induced pluripotent stem cell (iPSC) is considered a promising resource for restorative cell therapy in clinical areas. While fully reprogrammed iPSCs are similar to embryonic stem cells, iPSCs could be derived from the patient's own cells (autologous), which avoids the immune rejection activities. Recent advances have demonstrated that iPSCs could be generated from human fibroblasts using only four transcription factors: OCT4, SOX2, CMYC, and KLF4. However, the limitations of reprogramming technologies include low efficiency, slow kinetics, transgene integration and residual expression. Surprisingly, adult stem cells from human endometrium (endometrial stem cells; EnSCs) express OCT4 and KLF4 pluripotency factors. On the other hand, small molecule inhibitors of specific signaling pathways such as thiazovivin have been used in various aspects of iPSC generation and maintenance. Thiazovivin is a selective small molecule that directly targets Rho-associated kinase (ROCK) and increases expression of pluripotency factors. The process using thiazovivin could be easier, faster and more cost effective than transgene integration into somatic cells. So reprogramming of OCT4 and KLF4 expressing EnSCs by a ROCK inhibitor, thiazovivin, could result in producing more efficient iPSCs compared with fibroblasts or conventional somatic cells without integration any transgene and retroviral vector.


Subject(s)
Adult Stem Cells/drug effects , Cell Dedifferentiation/drug effects , Cellular Reprogramming/drug effects , Induced Pluripotent Stem Cells/physiology , Pyrimidines/pharmacology , Stromal Cells/drug effects , Thiazoles/pharmacology , Adult , Adult Stem Cells/physiology , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Kruppel-Like Factor 4 , Stromal Cells/physiology , rho-Associated Kinases/antagonists & inhibitors
14.
Neural Regen Res ; 9(8): 845-50, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-25206899

ABSTRACT

Several studies have demonstrated that selective serotonin reuptake inhibitor antidepressants can promote neuronal cell proliferation and enhance neuroplasticity both in vitro and in vivo. It is hypothesized that citalopram, a selective serotonin reuptake inhibitor, can promote the neuronal differentiation of adult bone marrow mesenchymal stem cells. Citalopram strongly enhanced neuronal characteristics of the cells derived from bone marrow mesenchymal stem cells. The rate of cell death was decreased in citalopram-treated bone marrow mesenchymal stem cells than in control cells in neurobasal medium. In addition, the cumulative population doubling level of the citalopram-treated cells was significantly increased compared to that of control cells. Also BrdU incorporation was elevated in citalopram-treated cells. These findings suggest that citalopram can improve the neuronal-like cell differentiation of bone marrow mesenchymal stem cells by increasing cell proliferation and survival while maintaining their neuronal characteristics.

15.
J Biol Eng ; 8: 20, 2014.
Article in English | MEDLINE | ID: mdl-25097665

ABSTRACT

First described in 2004, endometrial stem cells (EnSCs) are adult stem cells isolated from the endometrial tissue. EnSCs comprise of a population of epithelial stem cells, mesenchymal stem cells, and side population stem cells. When secreted in the menstrual blood, they are termed menstrual stem cells or endometrial regenerative cells. Mounting evidence suggests that EnSCs can be utilized in regenerative medicine. EnSCs can be used as immuno-modulatory agents to attenuate inflammation, are implicated in angiogenesis and vascularization during tissue regeneration, and can also be reprogrammed into induced pluripotent stem cells. Furthermore, EnSCs can be used in tissue engineering applications and there are several clinical trials currently in place to ascertain the therapeutic potential of EnSCs. This review highlights the progress made in EnSC research, describing their mesodermal, ectodermal, and endodermal potentials both in vitro and in vivo.

16.
Cell Biol Int ; 38(8): 901-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24715678

ABSTRACT

An increase in the number of viable in vitro differentiated neuronal cells is important for their use in clinics. A proportion of differentiated cells lose their viability before being used, and therefore we decided to use a pharmacological agent, sertraline, to increase neural cell differentiation and their survival. Purified endometrial stem cells (EnSCs) were examined for neuronal and glial cell specific markers after retinoic acid (RA) and sertraline treatment via RT-PCR, immunocytochemistry and Western blot analysis. The survival of differentiated cells was measured by MTT assay and the frequency of apoptosis, demonstrated by caspase-3-like activity. EnSCs were differentiated into neuronal cells after RA induction. Sertraline increased neuronal cell differentiation by 1.2-fold and their survival by 1.4-fold, and decreased from glial cell differentiation significantly. The findings indicate that sertraline could be used to improve the in vitro differentiation process of stem cells into neuronal cells, and may be involved in regenerative pharmacology in future.


Subject(s)
Cell Survival/drug effects , Mesenchymal Stem Cells/physiology , Neuroglia/physiology , Neurons/physiology , Sertraline/pharmacology , Tretinoin/pharmacology , Antigens, Differentiation/metabolism , Apoptosis/drug effects , Cell Differentiation , Cells, Cultured , Endometrium/cytology , Female , Humans , Neuroglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology
17.
PLoS One ; 8(11): e79907, 2013.
Article in English | MEDLINE | ID: mdl-24278212

ABSTRACT

Using phages is a novel field of cancer therapy and phage nanobioparticles (NBPs) such as λ phage could be modified to deliver and express genetic cassettes into eukaryotic cells safely in contrast with animal viruses. Apoptin, a protein from chicken anemia virus (CAV) has the ability to specifically induce apoptosis only in carcinoma cells. We presented a safe method of breast tumor therapy via the apoptin expressing λ NBPs. Here, we constructed a λ ZAP-CMV-apoptin recombinant NBP and investigated the effectiveness of its apoptotic activity on BT-474, MDA-MB-361, SKBR-3, UACC-812 and ZR-75 cell lines that over-expressing her-2 marker. Apoptosis was evaluated via annexin-V fluorescent iso-thiocyanate/propidium iodide staining, flow-cytometric method and TUNEL assay. Transfection with NBPs carrying λ ZAP-CMV-apoptin significantly inhibited growth of all the breast carcinoma cell lines in vitro. Also nude mice model implanted BT-474 human breast tumor was successfully responded to the systemic and local injection of untargeted recombinant λ NBPs. The results presented here reveal important features of recombinant λ nanobioparticles to serve as safe delivery and expression platform for human cancer therapy.


Subject(s)
Bacteriophage lambda/genetics , Breast Neoplasms/pathology , Capsid Proteins/genetics , Cell Division/genetics , Nanoparticles , Animals , Antineoplastic Agents , Female , Flow Cytometry , Humans , Mice , Mice, Nude , Recombinant Proteins/genetics
18.
BJU Int ; 112(6): 854-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24028767

ABSTRACT

OBJECTIVE: To investigate manufacturing smooth muscle cells (SMCs) for regenerative bladder reconstruction from differentiation of endometrial stem cells (EnSCs), as the recent discovery of EnSCs from the lining of women's uteri, opens up the possibility of using these cells for tissue engineering applications, such as building up natural tissue to repair prolapsed pelvic floors as well as building urinary bladder wall. MATERIALS AND METHODS: Human EnSCs that were positive for cluster of differentiation 146 (CD146), CD105 and CD90 were isolated and cultured in Dulbecco's modified Eagle/F12 medium supplemented with myogenic growth factors. The myogenic factors included: transforming growth factor ß, platelet-derived growth factor, hepatocyte growth factor and vascular endothelial growth factor. Differentiated SMCs on bioabsorbable polyethylene-glycol and collagen hydrogels were checked for SMC markers by real-time reverse-transcriptase polymerase chain reaction (RT-PCR), western blot (WB) and immunocytochemistry (ICC) analyses. RESULTS: Histology confirmed the growth of SMCs in the hydrogel matrices. The myogenic growth factors decreased the proliferation rate of EnSCs, but they differentiated the human EnSCs into SMCs more efficiently on hydrogel matrices and expressed specific SMC markers including α-smooth muscle actin, desmin, vinculin and calponin in RT-PCR, WB and ICC experiments. The survival rate of cultures on the hydrogel-coated matrices was significantly higher than uncoated cultures. CONCLUSIONS: Human EnSCs were successfully differentiated into SMCs, using hydrogels as scaffold. EnSCs may be used for autologous bladder wall regeneration without any immunological complications in women. Currently work is in progress using bioabsorbable nanocomposite materials as EnSC scaffolds for developing urinary bladder wall tissue.


Subject(s)
Endometrium/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Stem Cells/cytology , Tissue Engineering/methods , Actins/biosynthesis , Actins/genetics , Adult , Biopsy , Blotting, Western , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Endometrium/metabolism , Female , Gene Expression Regulation , Humans , Microscopy, Electron, Scanning , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Urinary Bladder/cytology
19.
Biomed Res Int ; 2013: 582526, 2013.
Article in English | MEDLINE | ID: mdl-24024202

ABSTRACT

Fluoxetine (FLX) is a selective serotonin reuptake inhibitor (SSRI). Its action is possibly through an increase in neural cell survival. The mechanism of improved survival rate of neurons by FLX may relate to the overexpression of some kinases such as Akt protein. Akt1 (a serine/threonine kinase) plays a key role in the modulation of cell proliferation and survival. Our study evaluated the effects of FLX on mesenchymal stem cell (MSC) fate and Akt1 phosphorylation levels in MSCs. Evaluation tests included reverse transcriptase polymerase chain reaction, western blot, and immunocytochemistry assays. Nestin, MAP-2, and ß-tubulin were detected after neurogenesis as neural markers. Ten µ M of FLX upregulated phosphorylation of Akt1 protein in induced hEnSC significantly. Also FLX did increase viability of these MSCs. Continuous FLX treatment after neurogenesis elevated the survival rate of differentiated neural cells probably by enhanced induction of Akt1 phosphorylation. This study addresses a novel role of FLX in neurogenesis and differentiated neural cell survival that may contribute to explaining the therapeutic action of fluoxetine in regenerative pharmacology.


Subject(s)
Fluoxetine/administration & dosage , Gene Expression Regulation, Developmental/drug effects , Mesenchymal Stem Cells/cytology , Proto-Oncogene Proteins c-akt/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mesenchymal Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/cytology , Neurons/drug effects , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Regenerative Medicine , Serotonin/metabolism , Up-Regulation
20.
Adv Pharmacol Sci ; 2013: 506191, 2013.
Article in English | MEDLINE | ID: mdl-24381588

ABSTRACT

Due to the increasing cases of neurodegenerative diseases in recent years, the eventual goal of nerve repair is very important. One approach for achieving a neuronal cell induction is by regenerative pharmacology. Nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) are neurotrophins that play roles in neuronal development, differentiation, and protection. On the other hand, dehydroepiandrosterone (DHEA) is a neurosteroid which has multiple actions in the nervous system. DHEA could be an important agent in regenerative pharmacology for neuronal differentiation during tissue regeneration. In this study, we investigated the possible role of DHEA to modulate NGF and BDNF production. The in vivo level of neurotrophins expression was demonstrated by ELISA in rat harvested brain cortex. Also neurotrophins expression after DHEA treatment was revealed by the increased neurite extension, immunostaining, and BrdU labeling in rats. Anti-NGF and anti-BDNF antibodies were used as suppressive agents on neurogenesis. The results showed that NGF and BDNF are overproduced after DHEA treatment but there is not any overexpression for NT-3 and NT-4. Also DHEA increased neurite extension and neural cell proliferation significantly. Overall, DHEA might induce NGF and BDNF neurotrophins overproduction in cortical neurons which promotes neural cell protection, survival, and proliferation.

SELECTION OF CITATIONS
SEARCH DETAIL
...