Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Biomedicines ; 12(6)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38927582

ABSTRACT

BACKGROUND: Mesenchymal stromal/stem cells (MSCs) play a critical role in wound healing. Corlicyte® is an MSC product derived from allogeneic umbilical cord tissue donated under an institutional review board-approved protocol and processed in accordance with section 501(a)(2)(B) of the Federal Food, Drug, and Cosmetic Act. This open-label phase 1 trial was performed under a United States Food and Drug Administration Investigational New Drug Application to establish the safety and tolerability of Corlicyte® in patients with diabetes and chronic diabetic foot ulcer (DFU). METHODS: Escalating doses were applied topically twice a week for up to 8 weeks after ulcer debridement, wound photography, and measurement. Subjects were followed for 4 weeks after the treatment phase. Adverse events were assessed at every visit. RESULTS: Nine subjects in 2 dosing cohorts completed the trial. No subjects experienced a serious adverse reaction to Corlicyte® or the development of anti-human leukocyte antigen (HLA) antibodies. Sixty percentage of subjects in the lower dose cohort experienced ulcer closure by Day 70 of follow-up, while the mean ulcer size was reduced by 54-67% in the other subjects. CONCLUSIONS: Topical administration of Corlicyte®, a novel biologic therapy consisting of allogeneic umbilical cord lining MSCs, appeared safe and tolerable and resulted in a significant decrease in ulcer area, demonstrating its potential as a therapy for healing of chronic DFU.

2.
Int J Mol Sci ; 23(16)2022 Aug 10.
Article in English | MEDLINE | ID: mdl-36012184

ABSTRACT

Human umbilical cord lining epithelial cells [CLECs) are naïve in nature and can be ethically recovered from cords that are routinely discarded. The success of using oral mucosal epithelial cells for cornea defects hints at the feasibility of treating cutaneous wounds using non-native CLECs. Herein, we characterized CLECs using flow cytometry (FC) and skin organotypic cultures in direct comparison with skin keratinocytes (KCs). This was followed by wound healing study to compare the effects of CLEC application and the traditional use of human skin allografts (HSGs) in a porcine wound model. While CLECs were found to express all the epidermal cell markers probed, the major difference between CLECs and KCs lies in the level of expression (in FC analysis) as well as in the location of expression (of the epithelium in organotypic cultures) of some of the basal cell markers probed. On the pig wounds, CLEC application promoted accelerated healing with no adverse reaction compared to HSG use. Though CLECs, like HSGs, elicited high levels of local and systemic immune responses in the animals during the first week, these effects were tapered off more quickly in the CLEC-treated group. Overall, the in vivo porcine data point to the potential of CLECs as a non-native and safe source of cells to treat cutaneous wounds.


Subject(s)
Umbilical Cord , Wound Healing , Animals , Epithelial Cells/metabolism , Humans , Keratinocytes , Skin/metabolism , Swine
3.
Int J Mol Sci ; 24(1)2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36613807

ABSTRACT

To determine the therapeutic efficacy of human umbilical cord lining mesenchymal stromal cells (CL-MSCs) (US Patent number 9,737,568) in lupus-prone MRL/lpr (Faslpr) mice and elucidate its working mechanisms. A total of 4 doses of (20-25) × 106 cells/kg of CL-MSCs was given to 16-week-old female Faslpr mice by intraperitoneal injection. Three subsequent doses were given on 17 weeks, 18 weeks, and 22 weeks, respectively. Six-week-old Faslpr mice were used as disease pre-onset controls. Mice were monitored for 10 weeks. Mouse kidney function was evaluated by examining complement component 3 (C3) deposition, urinary albumin-to-creatinine ratio (ACR), and lupus nephritis (LN) activity and chronicity. Working mechanisms were elucidated by flow cytometry, Luminex/ELISA (detection of anti-dsDNA and isotype antibodies), and RNA sequencing. CL-MSCs improved mice survival and kidney function by reducing LN activity and chronicity and lymphocyte infiltration over 10 weeks. CL-MSCs also reduced urinary ACR, renal complement C3 deposition, anti-dsDNA, and isotype antibodies that include IgA, IgG1, IgG2a, IgG2b, and IgM. Immune and cytokine profiling demonstrated that CL-MSCs dampened inflammation by suppressing splenic neutrophils and monocytes/macrophages, reducing plasma IL-6, IL-12, and CXCL1 and stabilizing plasma interferon-γ and TNF-α. RNA sequencing further showed that CL-MSCs mediated immunomodulation via concerted action of pro-proinflammatory cytokine-induced chemokines and production of nitric oxide in macrophages. CL-MSCs may provide a novel myeloid (neutrophils and monocytes/macrophages)-targeting therapy for SLE.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Mesenchymal Stem Cells , Female , Humans , Animals , Mice , Mice, Inbred MRL lpr , Kidney/metabolism , Cytokines/therapeutic use , Immunoglobulin G/therapeutic use , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism , Lupus Erythematosus, Systemic/therapy
4.
Cell Transplant ; 29: 963689719896559, 2020.
Article in English | MEDLINE | ID: mdl-32166974

ABSTRACT

We investigated the safety of using umbilical cord-lining stem cells for liver regeneration and tested a novel method for stem cell delivery. Stem cells are known by their ability to repair damaged tissues and have the potential to be used as regenerative therapies. The umbilical cord's outer lining membrane is known to be a promising source of multipotent stem cells and can be cultivated in an epithelial cell growth medium to produce cell populations which possess the properties of both epithelial cells and embryonic stem cells-termed cord-lining epithelial cells (CLEC). Hepatocytes are epithelial cells of the liver and their proliferation upon injury is the main mechanism in restoring the liver. Earlier studies conducted showed CLEC can be differentiated into functioning hepatocyte-like cells (HLC) and can survive in immunologically competent specimens. In this study, we chose a porcine model to investigate CLEC as a treatment modality for liver failure. We selected 16 immune competent Yorkshire-Dutch Landrace pigs, with a mean weight of 40.5 kg, for this study. We performed a 50% hepatectomy to simulate the liver insufficient disease model. After the surgery, four pigs were transplanted with a saline scaffold while seven pigs were transplanted with a HLC scaffold. Five pigs died on the surgical table and were omitted from the study analysis. This study addressed the safety of transplanting human CLEC in a large animal model. The transplant interfaces were evaluated and no signs of cellular rejection were observed in both groups.


Subject(s)
Epithelial Cells/cytology , Liver Regeneration/physiology , Mesenchymal Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Cells, Cultured , Epithelial Cells/physiology , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Stem Cells/physiology , Swine
5.
Front Cell Dev Biol ; 8: 596170, 2020.
Article in English | MEDLINE | ID: mdl-33763413

ABSTRACT

Purpose: We investigated the use of human Cord Lining Mesenchymal Stem Cells (CL-MSCs) (US Patent number 9,737,568), in a rabbit hindlimb ischemia model, and evaluated their potential in stimulating neovascularization. Allogenic human CL- MSCs could potentially be used to treat patients with lower limb ischemia and non-healing wounds. Methods: Twenty rabbits were divided into two separate groups. We created a hindlimb ischemia model surgically. At 21 and 49 days post-operatively, animals in the treatment group were injected with CL-MSCs (500,000 cells per 0.2 ml on each site) at 10 different sites (Quadriceps- 4 sites, Hamstrings- 4 sites and Calf--2 sites) in the hindlimb muscles. The control group received only saline injection to the corresponding sites at the same time point as the treatment group. We then evaluated the effects of treatment on neovascularization by angiography, laser doppler perfusion imaging, as well as by histology. We evaluated the tissue samples for any signs of local immune reaction to the cell implantation. We also observed the rabbit clinically for any adverse effects after treatment. Results: We found a higher number of CD31 positive cells in the treatment group, with a greater number of capillaries found in the treated muscles. The Rectus Femoris demonstrated a median vessel count/muscle fiber of 0.121 for the treatment group, compared to 0.076 in the control group (median difference 0.04; 95% CI 0.001-0.11; p = 0.041). The Gastrocnemius demonstrated a median vessel count/muscle fiber of 0.175 for the treatment group, compared to 0.089 in the control group (median difference 0.087; 95% CI -0.006 to 0.234; p = 0.07). Blood perfusion quantification through Laser Doppler Perfusion Imaging (LDPI) also demonstrated a non-statistically significant increase in perfusion in favor of the treatment group. CL-MSCs demonstrated no toxicity associated morbidity and minimal local immune reaction to implantation. Conclusion: CL-MSCs have a positive effect on angiogenesis in a rabbit hindlimb ischemia model. This preliminary data is encouraging and paves the way for future large animal studies or for clinical trials.

6.
Methods Mol Biol ; 1867: 141-164, 2018.
Article in English | MEDLINE | ID: mdl-30155821

ABSTRACT

Iatrogenic adverse events in clinical trials of retroviral vector-mediated gene-corrected cells have prioritized the urgent need for more comprehensive and stringent assessment of potentially genotoxic off-target alterations and the biosafety of cells intended for therapeutic applications. Genome editing tools such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced palindromic repeats (CRISPR)-Cas9 nuclease systems are being investigated as safer and efficient alternatives for site-directed genome modification. Using site-specific integration into the AAVS1 locus of primary human cells as an example, we present an integrated approach to multimodal investigation of off-target alterations and an evaluation of potential genotoxicity induced by ZFN-mediated integration of a therapeutic transgene.


Subject(s)
DNA Damage , Epithelial Cells/cytology , Gene Editing , Genetic Engineering/methods , Transgenes , Umbilical Cord/cytology , Zinc Finger Nucleases/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Genetic Vectors , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Recombination, Genetic , Transcriptome , Umbilical Cord/metabolism , Zinc Finger Nucleases/genetics
7.
Mol Ther ; 24(3): 607-19, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26689265

ABSTRACT

Costly coagulation factor VIII (FVIII) replacement therapy is a barrier to optimal clinical management of hemophilia A. Therapy using FVIII-secreting autologous primary cells is potentially efficacious and more affordable. Zinc finger nucleases (ZFN) mediate transgene integration into the AAVS1 locus but comprehensive evaluation of off-target genome effects is currently lacking. In light of serious adverse effects in clinical trials which employed genome-integrating viral vectors, this study evaluated potential genotoxicity of ZFN-mediated transgenesis using different techniques. We employed deep sequencing of predicted off-target sites, copy number analysis, whole-genome sequencing, and RNA-seq in primary human umbilical cord-lining epithelial cells (CLECs) with AAVS1 ZFN-mediated FVIII transgene integration. We combined molecular features to enhance the accuracy and activity of ZFN-mediated transgenesis. Our data showed a low frequency of ZFN-associated indels, no detectable off-target transgene integrations or chromosomal rearrangements. ZFN-modified CLECs had very few dysregulated transcripts and no evidence of activated oncogenic pathways. We also showed AAVS1 ZFN activity and durable FVIII transgene secretion in primary human dermal fibroblasts, bone marrow- and adipose tissue-derived stromal cells. Our study suggests that, with close attention to the molecular design of genome-modifying constructs, AAVS1 ZFN-mediated FVIII integration in several primary human cell types may be safe and efficacious.


Subject(s)
Endonucleases/metabolism , Factor VIII/genetics , Genome-Wide Association Study , Mutagenesis, Insertional , Zinc Fingers , Binding Sites , Factor VIII/metabolism , Gene Expression , Gene Targeting , Gene Transfer Techniques , Genetic Vectors/genetics , High-Throughput Nucleotide Sequencing , Humans , K562 Cells , Protein Binding , Transgenes
8.
Invest Ophthalmol Vis Sci ; 56(8): 4697-704, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26207305

ABSTRACT

PURPOSE: To determine the effectiveness of human umbilical cord-derived mucin-expressing cord lining epithelial cells (CLEC-muc) as feeder cells in a coculture system for the cultivation of human limbal stem cells. METHODS: Human CLEC-muc were cultured in PTTe-1 medium and treated with mitomycin C to arrest their growth to make the feeder layer. Single-cell suspension of limbal cells was prepared from corneal rim collected from the Singapore Eye Bank. Limbal cells were cultured in a coculture system with CLEC-muc as well as 3T3 cells as feeder layer. We compared the colony-forming efficiency and cell morphology of the limbal cells cultured in the two different feeder layers. We also compared the expression level of several putative limbal stem cell markers, such as HES1, ABCG2, ΔNP63, and BMI1, in the cultured limbal cells by immunostaining and quantitative (q)RT-PCR. Expression of cytokeratins CK14, CK15, CK19, CK3, and CK4 was further compared. RESULTS: Human limbal epithelial cells cultured in both types of feeder layers showed comparable cell morphology and colony-forming efficiency. These cells exhibited a similar expression pattern of HES1, ABCG2, ΔNP63, BMI1, CK14, CK15, CK19, and CK3 as detected by immunostaining and PCR. CONCLUSIONS: Human CLEC-muc may be a suitable alternative to conventional mouse 3T3 feeder cells, which may reduce the risk of zoonotic infection.


Subject(s)
Epithelium, Corneal/cytology , Limbus Corneae/cytology , Stem Cells/cytology , Umbilical Cord/cytology , Biomarkers/metabolism , Cell Count , Cell Line , Cell Proliferation , Coculture Techniques , DNA/genetics , Epithelium, Corneal/growth & development , Epithelium, Corneal/metabolism , Feeder Cells , Gene Expression Regulation , Gene Expression Regulation, Developmental , Humans , Keratins/biosynthesis , Keratins/genetics , Limbus Corneae/growth & development , Limbus Corneae/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
9.
Adv Wound Care (New Rochelle) ; 4(2): 110-118, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25713753

ABSTRACT

Objective: Nanofibers for tissue scaffolding and wound dressings hold great potential in realizing enhanced healing of wounds in comparison with conventional counterparts. Previously, we demonstrated good fibroblast adherence and growth on a newly developed scaffold, Tegaderm™-Nanofiber (TG-NF), made from poly ɛ-caprolactone (PCL)/gelatin nanofibers electrospun onto Tegaderm (TG). The purpose of this study is to evaluate the performance and safety of TG-NF dressings in partial-thickness wound in a pig healing model. Approach: To evaluate the rate of reepithelialization, control TG, human dermal fibroblast-seeded TG-NF(+) and -unseeded TG-NF(-) were randomly dressed onto 80 partial-thickness burns created on four female and four male pigs. Wound inspections and dressings were done after burns on day 7, 14, 21, and 28. On day 28, full-thickness biopsies were taken for histopathological evaluation by Masson-Trichrome staining for collagen and hematoxylin-eosin staining for cell counting. Results: No infection and severe inflammation were recorded. Wounds treated with TG-NF(+) reepithelialized significantly faster than TG-NF(-) and control. Wound site inflammatory responses to study groups were similar as total cell counts on granulation tissues show no significant differences. Most of the wounds completely reepithelialized by day 28, except for two wounds in control and TG-NF(-). A higher collagen coverage was also recorded in the granulation tissues treated with TG-NF(+). Innovation and Conclusion: With better reepithelialization achieved by TG-NF(+) and similar rates of wound closure by TG-NF(-) and control, and the absence of elevated inflammatory responses to TG-NF constructs, TG-NF constructs are safe and demonstrated good healing potentials that are comparable to Tegaderm.

10.
Cell Transplant ; 23(4-5): 497-503, 2014.
Article in English | MEDLINE | ID: mdl-24636188

ABSTRACT

Intense scientific research over the past two decades has yielded much knowledge about embryonic stem cells, mesenchymal stem cells from bone marrow, as well as epithelial stem cells from the skin and cornea. However, the billions of dollars spent in this research have not overcome the fundamental difficulties intrinsic to these stem cell strains related to ethics (embryonic stem cells), as well as to technical issues such as accessibility, ease of cell selection and cultivation, and expansion/mass production, while maintaining consistency of cell stemness (all of the stem cell strains already mentioned). Overcoming these technical hurdles has made stem cell technology expensive and any potential translational products unaffordable for most patients. Commercialization efforts have been rendered unfeasible by this high cost. Advanced biomedical research is on the rise in Asia, and new innovations have started to overcome these challenges. The Nobel Prize-winning Japanese development of iPSCs has effectively introduced a possible replacement for embryonic stem cells. For non-embryonic stem cells, cord lining stem cells (CLSCs) have overcome the preexisting difficulties inherent to mesenchymal stem cells from the bone marrow as well as epithelial stem cells from the skin and cornea, offering a realistic, practical, and affordable alternative for tissue repair and regeneration. This novel CLSC technology was developed in Singapore in 2004 and has 22 international patents granted to date, including those from the US and UK. CLSCs are derived from the umbilical cord outer lining membrane (usually regarded as medical waste) and is therefore free from ethical dilemmas related to its collection. The large quantity of umbilical cord lining membrane that can be collected translates to billions of stem cells that can be grown in primary stem cell culture and therefore very rapid and inexpensive cell cultivation and expansion for clinical translational therapies. Both mesenchymal and epithelial stem cells can be isolated from the umbilical cord lining membrane, usefully regenerating not only mesenchymal tissue, such as bone, cartilage, and cardiac and striated muscle, but also epithelial tissue, such as skin, cornea, and liver. Both mesenchymal and epithelial CLSCs are immune privileged and resist rejection. Clinically, CLSCs have proved effective in the treatment of difficult-to-heal human wounds, such as diabetic ulcers, recalcitrant chronic wounds, and even persistent epithelial defects of the cornea. Heart and liver regeneration has been shown to be successful in animal studies and await human trials. CLSCs have also been shown to be an effective feeder layer for cord blood hematopoietic stem cells and, more recently, has been recognized as an abundant and high-quality source of cells for iPSC production. Banking of CLSCs by cord blood banks in both private and public settings is now available in many countries, so that individuals may have their personal stores of CLSCs for future translational applications for both themselves and their families. Cord lining stem cells are strongly positioned to be the future of cell therapy and regenerative medicine.


Subject(s)
Epithelial Cells/cytology , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Animals , Antigens, CD/metabolism , Cell Differentiation , Epithelial Cells/metabolism , Heart Diseases/therapy , Humans , Mesenchymal Stem Cell Transplantation , Regenerative Medicine
11.
Assay Drug Dev Technol ; 11(2): 130-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23113625

ABSTRACT

The primary hepatocyte is the best benchmark for drug biotransformation studies. However, due to the severe shortage of primary hepatocytes, there is a need for alternative reliable cell source. This study aims to isolate multipotent epithelial cells from the umbilical cord, differentiate these cells into hepatocyte-like cells (HLCs), and investigate the potential of using the differentiated cells for in vitro drug metabolism model. Human umbilical cord lining epithelial cells (UCLECs) were subjected to hepatic induction over a period of 28 days. HepG2 and cryopreserved human hepatocytes were used as control. Immunohistological staining was carried out for α-fetoprotein (AFP), albumin, cytokeratin 18 (CK18), and 19 (CK19). Glycogen storage ability was assessed through periodic acid-Schiff stain. Reverse transcription polymerase chain reaction was performed to examine gene expression of hepatic nuclear factor 4α (HNF4α) and cytochrome P450 isozymes 1A2, 2C9, 2D6, and 3A4. Ultra-performance liquid chromatography tandem mass spectrometry (UPLC/MS/MS) was utilized to analyze functional metabolic ability of the HLCs, where CYP3A4 was chosen as the study focus and testosterone as the drug substrate. After 28 days of induction, the fibroblastic phenotype of UCLECs changed to rotund polygonal shape resembling that of hepatocytes. Protein expression of AFP and CK19 was negative, while albumin and CK18 expression was upregulated. Gene expression of HNF4α, CYP1A2, CYP2D6, and CYP3A4 was observed but not for CYP2C9. After 4 h of incubation with testosterone, UPLC/MS/MS detected 2α-, 6ß-, 15ß-, and 16ß-hydroxytestosterone. UCLECs are able to differentiate into HLCs that express liver-specific markers, and have functional metabolic capabilities.


Subject(s)
Batch Cell Culture Techniques/methods , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Umbilical Cord/cytology , Umbilical Cord/physiology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Proteome/metabolism
12.
J Dermatol Sci ; 64(3): 199-209, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22005028

ABSTRACT

BACKGROUND: Current evidence suggests the potential role of Wnt signalling in keloids pathogenesis but such literature remains scanty. We hypothesize that Wnt signalling is upregulated in keloid fibroblasts (KFs) and this promotes cellular growth, migration and extracellular matrix (ECM) production in such fibroblasts. OBJECTIVES: To verify the downregulation of secreted frizzled-related protein 1 (SFRP1), a Wnt inhibitor and test KFs sensitivity to Wnt3a treatment compared to NFs in terms of activation of Wnt/ß-catenin, cellular growth, migration and ECM expressions. Next, to investigate if ectopic expression of SFRP1 and treatment of quercetin in KFs can reverse their phenotypes. METHODS: Quantitative Real-time PCR and western blotting were used to verify SFRP1 expression in NFs and KFs. The fibroblasts were tested with Wnt3a conditioned media and its effects were tested for (1) the cells' sensitivity to direct Wnt signalling via the activation of TCF reporter assay and protein expression of ß-catenin, (2) cellular growth, (3) cell migration and (4) expressions of ECM components. Finally KFs were stably transduced with SFRP1 and treated with 2 doses of quercetin. RESULTS: Lower levels of SFRP1 were confirmed at mRNA and protein levels in KFs which partly explained their sensitivity to Wnt3a treatment in terms of higher Wnt activation, cellular growth and fibronectin expression. Interestingly, Wnt3a did not promote higher cell migration rate and increase in collagen I expression. Ectopic expression of SFRP1 and quercetin treatment was able to mitigate Wnt3a-mediated phenotype of KFs. CONCLUSIONS: Using SFRP1 or inhibitors of Wnt signalling might be one of the therapeutic solutions to treat keloid scarring.


Subject(s)
Cell Proliferation , Fibroblasts/metabolism , Fibronectins/metabolism , Keloid/metabolism , Wnt3A Protein/metabolism , Adolescent , Adult , Blotting, Western , Case-Control Studies , Cell Movement , Cell Proliferation/drug effects , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Keloid/genetics , Keloid/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phenotype , Quercetin/pharmacology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Time Factors , Transfection , Young Adult , beta Catenin/metabolism
13.
Theor Biol Med Model ; 8: 13, 2011 May 02.
Article in English | MEDLINE | ID: mdl-21535890

ABSTRACT

BACKGROUND: Keloids are protrusive claw-like scars that have a propensity to recur even after surgery, and its molecular etiology remains elusive. The goal of reverse engineering is to infer gene networks from observational data, thus providing insight into the inner workings of a cell. However, most attempts at modeling biological networks have been done using simulated data. This study aims to highlight some of the issues involved in working with experimental data, and at the same time gain some insights into the transcriptional regulatory mechanism present in keloid fibroblasts. METHODS: Microarray data from our previous study was combined with microarray data obtained from the literature as well as new microarray data generated by our group. For the physical approach, we used the fREDUCE algorithm for correlating expression values to binding motifs. For the influence approach, we compared the Bayesian algorithm BANJO with the information theoretic method ARACNE in terms of performance in recovering known influence networks obtained from the KEGG database. In addition, we also compared the performance of different normalization methods as well as different types of gene networks. RESULTS: Using the physical approach, we found consensus sequences that were active in the keloid condition, as well as some sequences that were responsive to steroids, a commonly used treatment for keloids. From the influence approach, we found that BANJO was better at recovering the gene networks compared to ARACNE and that transcriptional networks were better suited for network recovery compared to cytokine-receptor interaction networks and intracellular signaling networks. We also found that the NFKB transcriptional network that was inferred from normal fibroblast data was more accurate compared to that inferred from keloid data, suggesting a more robust network in the keloid condition. CONCLUSIONS: Consensus sequences that were found from this study are possible transcription factor binding sites and could be explored for developing future keloid treatments or for improving the efficacy of current steroid treatments. We also found that the combination of the Bayesian algorithm, RMA normalization and transcriptional networks gave the best reconstruction results and this could serve as a guide for future influence approaches dealing with experimental data.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , Gene Regulatory Networks/genetics , Genetic Engineering/methods , Keloid/genetics , Algorithms , Binding Sites , Culture Media, Serum-Free , Databases, Genetic , Fibroblasts/drug effects , Gene Regulatory Networks/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Keloid/pathology , Receptors, Cytokine/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Steroids/pharmacology , Transcription Factors/metabolism , Transcription, Genetic/drug effects
14.
Stem Cell Rev Rep ; 7(4): 935-47, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21431286

ABSTRACT

The stem cells involved in renewal of the corneal epithelium are located in the basal region of the limbus, a narrow transition zone surrounding the cornea. In many ocular surface disorders loss of these stem cells results in partial or complete vision loss. Conventional corneal transplant in these patients is associated with dismal results. Stem cell transplantation offers new hope to such patients. The umbilical cord is emerging as an important source of stem cells that may have potential clinical applications. There are advantages to the use of umbilical cord stem cells as these cells are less immunogenic, non-tumorigenic, highly proliferative and ethically acceptable. In this study, we have confirmed the expression of several putative limbal stem cell markers such as HES1, ABCG2, BMI1, CK15 as well as cell adhesion-associated molecules INTEGRIN-α6, -α9, -ß1, COLLAGEN-IV and LAMININ in our recently characterized CLEC-muc population derived from human umbilical cord. Ex vivo expansion of these cells on a human amniotic membrane substrate formed a stratified cell sheet that similarly expresses some of these molecules as well as cornea-specific cytokeratins, CK3 and CK12. Transplantation of a bioengineered CLEC-muc sheet in limbal stem cell-deficient rabbit eyes resulted in regeneration of a smooth, clear corneal surface with phenotypic expression of the normal corneal-specific epithelial markers CK3, CK12 but not CK4 or CK1/10. Our results suggest that CLEC-muc is a novel stem cell that can be ex vivo expanded for corneal epithelial regeneration in the treatment of various eye diseases.


Subject(s)
Corneal Diseases/therapy , Regeneration , Stem Cells/cytology , Umbilical Cord/cytology , Amnion/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cells, Cultured , Corneal Diseases/metabolism , Epithelium, Corneal/injuries , Epithelium, Corneal/metabolism , Epithelium, Corneal/transplantation , Gene Expression Regulation , Humans , Immunohistochemistry , Limbus Corneae/injuries , Limbus Corneae/metabolism , Models, Animal , Phenotype , Rabbits , Stem Cell Transplantation , Stem Cells/metabolism , Tissue Engineering/methods , Transplantation, Heterologous
15.
Cell Transplant ; 20(11-12): 1827-41, 2011.
Article in English | MEDLINE | ID: mdl-21439131

ABSTRACT

In this study we describe the derivation and immunological characterization of a primary epithelial cell type from the human umbilical cord membrane. These cord lining epithelial cells (CLECs) expressed and/or secreted isoforms of the nonclassical human leukocyte antigen class I (HLA-1b) glycoproteins, HLA-G and E. Conditioned media from CLECs inhibited mitogen-stimulated T-lymphocyte responses, and in a mixed leukocyte reaction (MLR) assay, cocultured CLECs inhibited allogeneic responses with a concomitant reduction in proinflammatory cytokines. Using a transwell coculture system, it was demonstrated that these immunoregulatory effects were mediated by soluble factors secreted by CLECs, in a dose-dependent manner. Functional studies using HLA-G blocking antibody showed that the effects of CLEC-secreted products could be inhibited, thus demonstrating a significant and important role for soluble HLA-G. In vivo, we show that transplanted CLECs could be maintained for extended periods in immunocompetent mice where xenorejection rapidly destroyed primary keratinocytes, a control human epithelial cell type. Additionally, CLECs delayed the rejection of keratinocytes and extended their survival when cotransplanted, indicating an ability to protect adjacent human cell types that would otherwise be rejected if transplanted alone. We also show that CLECs transduced with a modified human proinsulin gene were transplanted intraperitoneally into streptozotocin (STZ)-induced diabetic mice, resulting in significantly lower levels of serum glucose compared to control mice. This study has characterized the immunological properties of CLECs and tested a potential therapeutic application in the treatment of a type 1 diabetes mouse model.


Subject(s)
Epithelial Cells/metabolism , Fetal Blood/cytology , Animals , Blood Glucose/analysis , Cells, Cultured , Diabetes Mellitus, Experimental/therapy , Epithelial Cells/cytology , Epithelial Cells/transplantation , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/immunology , HLA-G Antigens/immunology , HLA-G Antigens/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/transplantation , Leukocytes, Mononuclear/cytology , Mice , Mice, SCID , Proinsulin/genetics , Proinsulin/metabolism , Transplantation, Heterologous , HLA-E Antigens
16.
Stem Cell Rev Rep ; 7(3): 624-38, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21181306

ABSTRACT

Umbilical cord tissue is gaining attention as a novel source of multipotent stem cells because it is easily obtainable, ethically acceptable and the cells are immunologically naïve. In this study, we have isolated and characterized a new cell type expressing MUCIN1 (CD227) from human umbilical cord lining which we termed MUCIN-expressing Cord Lining Epithelial Cell (CLEC-muc). We found that CLEC-muc is highly proliferative and had significant clonogenic ability. These cells express embryonic stem cell markers OCT-4, NANOG, SSEA-4, REX1 and SOX2. Despite the abundant expression of epithelial cell marker MUCIN1 and cytokeratins, this population is also positive to the mesenchymal stem cell (MSC) marker CD166. CLEC-muc is unique in p63 expression that shuttles from the cytoplasm to the nucleus over time in culture. To understand p63 regulation and function in CLEC-muc, cells were treated with BMP4, a potent morphogen that plays a role in epidermal differentiation via p63 upregulation in ES cell and subsequent analyses were done. We found that BMP4 does not alter cytoplasmic expression of p63 that promotes cell proliferation. However, it increases nuclear p63 expression together with several other epithelial-associated genes such as GATA3, JAGGED1, NOTCH1, HES1 and IKKα. BMP4 has also been found to weakly induce deltaNp63 expression in CLEC-muc. Our results suggest that CLEC-muc is a novel stem cell-like population that can be further differentiated by BMP4 to generate specific cell-types probably destined to form non-keratinized epithelia.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Membrane Proteins/metabolism , Mucin-1/metabolism , Stem Cells/cytology , Stem Cells/physiology , Umbilical Cord/cytology , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Proliferation , Cell Separation , Cells, Cultured , Female , Flow Cytometry , Humans , Pregnancy
17.
Exp Dermatol ; 19(11): 987-93, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20812968

ABSTRACT

The keloid fibroblast (KF) is known to have higher proliferative capacity than normal dermal fibroblast (NF). Metallothionein (MT), a metal-binding protein, has been reported to promote cell proliferation. In this study, we evaluated the expression of MT isoforms at the mRNA level in fetal bovine serum (FBS)-stimulated proliferating KF. Although the morphological appearance of NF and KF was similar when viewed under light, confocal and transmission electron microscopy, there was surprisingly a generally lower expression of MT isoforms in KF when compared with NF and also reduced MT staining in dermal fibroblasts of keloids as opposed to normal skin. Primary cultures of KF grown in 5% FBS or 10% FBS compared to without FBS demonstrated significantly higher proliferative activity and more abundant deposition of collagen. Contrary to expectation, MT-1A, -1F, -1G, -1X and -2A isoforms were significantly down-regulated in proliferating KF. Moreover, stimulating KF with TGF ß1, which is known to promote collagen synthesis and keloid formation, increased expression of Collagen 1A and 3A genes accompanied by reduction in MT-2A gene expression. Furthermore, down-regulation of the MT-2A gene in proliferating KF by siRNA-mediated silencing enhanced cell proliferation with concomitant up-regulation of the NF-κB gene and 10 of 13 other NF-κB pathway-related genes analysed but no alteration of the Collagen 1 and Collagen 3 gene expression. It would appear that down-regulation of MT isoforms in proliferating KF, in particular MT-2A, enhances keloidogenesis with the possible involvement of the NF-κB signalling pathway.


Subject(s)
Cell Proliferation , Collagen/metabolism , Fibroblasts/metabolism , Keloid/pathology , Metallothionein/metabolism , Protein Isoforms/metabolism , Cells, Cultured , Collagen/genetics , Culture Media, Serum-Free/pharmacology , Down-Regulation/genetics , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression/drug effects , Gene Expression/genetics , Humans , In Vitro Techniques , Keratinocytes/metabolism , Keratinocytes/pathology , Metallothionein/genetics , NF-kappa B/genetics , Protein Isoforms/genetics , RNA, Small Interfering/genetics , Serum/physiology , Signal Transduction/genetics , Skin/metabolism , Skin/pathology , Transforming Growth Factor beta1/pharmacology , Up-Regulation/genetics
18.
Mol Ther ; 18(7): 1346-56, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20424600

ABSTRACT

Biosafety and efficacy considerations that impede clinical application of gene therapy could be addressed by nonviral ex vivo cell therapy, utilizing transgenic cells that have been comprehensively pre-evaluated for genotoxic potential and transgene expression. We evaluated the genotoxic potential of phiC31 bacteriophage integrase-mediated transgene integration in cord-lining epithelial cells (CLECs) readily cultured from the outer membrane of human umbilical cords, by sequencing and mapping integration sites, spectral karyotyping, high-resolution genome copy number, transcriptome, and transgene copy number analyses and in vivo tumorigenicity. Of 44 independent integration events, <5% were exonic and 85% of modified cells had integrated

Subject(s)
Epithelial Cells/cytology , Transgenes/genetics , Umbilical Cord/cytology , Animals , Blotting, Western , Cells, Cultured , Electroporation , Epithelial Cells/metabolism , Factor VIII/genetics , Factor VIII/metabolism , Fluorescent Antibody Technique, Indirect , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
Ann Surg ; 246(5): 886-95, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17968183

ABSTRACT

OBJECTIVE: We examined the transcriptional response to serum stimulation as an in vitro model of wound healing in keloid fibroblasts to identify molecular mechanisms leading to their aberrant growth. SUMMARY BACKGROUND DATA: Keloids are proliferative dermal growths representing a pathologic wound healing response. Although several groups have shown increased expression of profibrotic factors in keloids, there is little known about why they are expressed at higher levels than normal. METHODS: Fibroblasts derived from keloids and normal scar were subjected to serum stimulation as an in vitro model to mimic a component of the wound microenvironment to examine differential gene expression in keloid derived fibroblasts versus normal human fibroblasts. A promoter analysis was performed to identify specific enhancers involved in mediating the differential response of connective tissue growth factor (CTGF, CCN2). Point mutations in the enhancers were performed to confirm their role. Finally, we examined activation of transcription factors known to bind the targeted enhancers. RESULTS: Transcription of CCN2 after serum stimulation was significantly higher in keloid versus normal fibroblasts. Promoter analysis demonstrates the fragment from -625/-140 conferred increased serum responsiveness. Mutational analysis showed an AP-1 and SMAD binding site were both necessary for serum responsiveness. Preventing activation of either transcriptional complex will block CCN2 transcription. Additional experiments suggest that a single complex that includes components of the AP-1 and SMAD binding complexes is responsible for transactivation in response to serum. The key difference between keloid and normal fibroblasts appears to be the degree of activation of c-Jun. CONCLUSIONS: We suggest that altered responsiveness to cellular stress, based upon current data using serum stimulation and past data on response to mechanical strain, is a key defect leading to keloid formation.


Subject(s)
Fibroblasts/physiology , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Keloid/metabolism , Serum/physiology , Smad Proteins, Receptor-Regulated/metabolism , Transcription Factor AP-1/metabolism , Adolescent , Adult , Binding Sites/physiology , Cell Culture Techniques , Connective Tissue Growth Factor , Cysteine-Rich Protein 61 , Humans , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Keloid/genetics , Keloid/pathology , RNA, Messenger/metabolism , Smad Proteins, Receptor-Regulated/genetics , Transcription Factor AP-1/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...