What is the Best Source of Stem Cells? Bone Marrow versus Adipose (Fat)? Part 2

This is a very important question. It does appear Adipose (Fat) is a better source of stem cells, in terms of quantity and percentage of key markers of viability, noted in below studies, than Bone Marrow. I did post about this last year in May. https://drcremers.com/2017/05/what-is-best-source-of-stem-cells.html

Since then there have been more studies. Though I could find none on the use of Bone Marrow versus Adipose derived Stem Cells for use in regenerating Meibomian Gland cells, there are good studies looking at the benefit of topical (drops only) use of adipose-derived stem cells to heal corneal issues faster than control groups. There are also good articles on adipose derived stem cells to heal lacrimal glands: coming in another post but initial articles are listed below **.

The first Reference below is on the use of Adipose Derived Stem Cells to support the growth of limbal stem cells.

The key finding in general is that adipose derived stem cells are generally safe in animals and humans for certain applications: not all. The complication rate is low for almost all types of injections I could find thus far except injection into the eye vitreous cavity where 3 patients have lost vision from this.

While the animal studies look very promising, there is no 100% guarantee stem cells will work for a particular condition.

Use of human adipose derived stem cells for injection into the orifice of the meibomian gland has not been done yet. It looks promising in general.

We hope to start our adipose derived stem cell injection into the orifice of the meibomian gland hopefully in the next few weeks, pending IRB approval. We appear to have received our IRB approval from Georgetown University’s Medical School for the meibomian gland disease issue I have posted about in kids using excessive computers.

Sandra Lora Cremers, MD, FACS

A side note about published studies:
1. I always like to look at which group did the study. Some countries have more rigorous criteria for research than others. Some groups are well known for the quality of their research. Groups from the US are in general more respected.
2. Higher impact factor journals are better usually. I have posted about this also before: Pubmed should have impact journal scores next to titles on the search options.
3. In general: It is best to look at articles that have a control group.
4. All studies need to be reproduced by another independent group before it can be verified.

References:
1.  With permission, I have posted the entire article of this paper.
They showed topical stem cell drops healed corneal wounds in mice faster than 100% autologous serum.

Notes:
1. Isolation, preparation, flow cytometry, immunofluorescence, and multilineage differentiation of ADSC have been reported in our previous studies [,,].
12. Beltrami A.P., Cesselli D., Bergamin N., Marcon P., Rigo S., Puppato E., D’Aurizio F., Verardo R., Piazza S., Pignatelli A., et al. Multipotent cells can be generated in vitro from several adult human organs (heart, liver, and bone marrow) Blood. 2007;

17. Zeppieri M., Salvetat M.L., Beltrami A.P., Cesselli D., Bergamin N., Russo R., Cavaliere F., Varano G.P., Alcalde I., Merayo J., et al. Human adipose-derived stem cells for the treatment of chemically burned rat cornea: Preliminary results. Curr. Eye Res. 2013;38:451–463. doi: 10.3109/02713683.2012.763100.[PubMed] [Cross Ref]

18. Ferro F., Spelat R., Falini G., Gallelli A., D’Aurizio F., Puppato E., Pandolfi M., Beltrami A.P., Cesselli D., Beltrami C.A., Ambesi-Impiombato F.S. Adipose tissue-derived stem cell in vitro differentiation in a three-dimensional dental bud structure. Am. J. Pathol. 2011;178:2299–2310. doi: 10.1016/j.ajpath.2011.01.055. [PMC free article] 

. 2017 Dec; 6(12): 115.
Published online 2017 Dec 5. doi:  10.3390/jcm6120115
PMCID: PMC5742804

Adipose Derived Stem Cells for Corneal Wound Healing after Laser Induced Corneal Lesions in Mice

*Correspondence: moc.liamtoh@ireippezkram; Tel.: +39-432-552-743; Fax: +39-432-552-741

1. Introduction

The cornea provides a protective barrier and clear optical pathway for the visual system. This avascular and transparent structure is composed of three layers, which include the outer most non-keratinized stratified epithelium, stroma, and single-layered endothelium. Most corneal diseases and trauma involve the epithelium and stroma and include immune disorders, chronic inflammation, infection, iatrogenic procedures (i.e., laser refractive surgery), etc. These ocular disorders tend to cause severe inflammation, persistent epithelium defects, neovascularization, conjunctivalization, persistent corneal opacities, and scarring, which can all lead to permanent vision loss [,,].
The most common intervention for restoring corneal clarity and function is full thickness penetrating or lamellar keratoplasty. However, there are great limitations related to little availability of donor tissue and costs and complications related to surgery (i.e., graft failure, chronic inflammation, numerous follow-up, etc.). Alternative surgical techniques include amniotic membrane transplantation and conjunctival limbal autograft from the healthy fellow eye, but surgical outcomes and limitations are similar to keratoplasty []. Studies have shown that autologous serum [] autologous plasma rich in growth factors [,] and other novel topical treatments [,] can be beneficial in patients with various ocular surface disorders due to inflammation, severe dryness, and persistent epithelial defects [,,]. However, long-term clinical efficacy multicenter data and standardized preparation protocols are lacking. Moreover, therapeutic effects tend to be superficial, limiting and symptomatic involving the corneal epithelial and not directed on lesion induced stromal scarring and damage.
In the past decade, stem cell therapy has been proposed in almost every branch of medicine, ranging from cardiology, neurology, plastic surgery, dentistry, etc. [,,,] Similar to bone marrow derived stem cells, numerous studies have shown that adipose derived stem cells (ADSC) are pluripotent and have the capability of differentiating into multiple mesodermal cell lineages expressing specific markers and proteins [,,]. The interest in ADSC has greatly increased considering that it is easy to obtain large quantities of autologous tissue in a rapid minimally invasive liposuction procedure that requires only local anesthesia. Moreover, ADSC are extremely more abundant in adipose tissue than MSC in bone marrow [].
Numerous studies based on ADSC obtained from processed human lipoaspirate to treat corneal lesions in various animal models have shown stem cells to be able to differentiate and produce corneal specific proteins, thus enhancing wound healing and maintaining transparency [], while providing inhibition of inflammation [] and angiogenesis []. ADSC can be placed in corneal stromal pockets. However, surgical manipulation of cells and of the host cornea can be potentially damaging and may not be of potential clinical use in a clinical setting.
Topical stem cell therapy may be of potential clinical use considering that ADSC are abundant, cost efficient, multipotent, non-invasive, and beneficial in various phases of wound healing including cellular remodeling, changes to tear composition, cell migration, proliferation, epithelial reattachment, and stromal remodeling. Our previous study reported the topical use of ADSC in chemically induced corneal lesions in rats []. These preliminary results showed that stem cell treated eyes had significantly smaller epithelial defects at each time point, with better and faster re-epithelization and less inflammatory response compared to other treatment arms. The ADSC group seemed to show improved corneal wound healing. However, a small number of animals with chemically induced lesions in a brief time were considered in the treatment and in the clinical assessment to provide a semi-quantitative assessment of data. The purpose of our current study was to assess a greater number of eyes over a longer time period using a reproducible laser-induced corneal wound in mice in addition to provide a comparison of epithelial repair, stromal haze, inflammation, and quantitative histological analysis between treatment groups.

2. Material and Methods

2.1. Animals

Forty black male mice C57BL/6 (30–40 g) purchased from Charles River Laboratories (Barcelona, Spain) were used in the experiments. Animals were housed with a 12-h light–dark cycle with ad libitum access to food and water. Animal care and experiments were carried out in accordance with the guidelines of the Spanish Ministry of Health for Animal Care (RD 53/2013) and European Commissions (2003/65/EC).
Prior to induction of lesion and treatments, mice were anesthetized by intraperitoneal injection of 80 mg/kg ketamine hydrochloride (Imalgène 500, Merial, Lyon, France) and 5 mg/kg xylazine hydrochloride (Rompun, Bayer HealthCare, Kiel, Germany). Topical anesthesia was induced by 0.4% oxibuprocain eye drops (Novesina, Novartis, Varese, Italy). Animals were euthanized with an overdose of sodium pentobarbital (Dolethal, Vétoquinol, Lure, France) and verified by cervical dislocation.

2.2. Isolation and Preparation of Adipose Derived Stem Cells

Human subcutaneous abdominal adipose tissue was obtained from healthy patients (aged 35–64 years) undergoing elective lipoaspiration surgery with informed oral and written consent under a protocol approved by the Institutional Review Board (IRB) of the University of Udine, in accordance with the guidelines of the Tenets of the Declaration of Helsinki. Patients were screened and resulted negative for HIV, hepatitis B and C virus, and syphilis.
Isolation, preparation, flow cytometry, immunofluorescence, and multilineage differentiation of ADSC have been reported in our previous studies [,,].
For the in vivo experiment, we employed ADSC obtained from a 35 year old female patient, at the third passage in culture, in their undifferentiated state.

2.3. Blood Serum

Human serum was prepared in accordance to the Azienda Ospedaliero Universitaria Santa Maria della Misericordia protocol []. In brief, 500 mL of whole blood from one healthy young male donor (45 years old) was collected into sterile 9 mL tubes after written informed consent. The patient was screened and resulted negative for HIV, hepatitis B and C virus, and syphilis. The containers were left standing in an upright position to ensure clotting for at least 30 min at room temperature, then centrifuged at 2000× g for 10 min. The supernatant serum was removed under sterile conditions in a laminar flow hood with sterile disposable syringes. The vials were frozen and left to thaw for 24 h before treatment. In accordance to our hospital protocols on clinical treatment for severe dry eye syndrome, we used undiluted whole serum.

2.4. Laser Induced Corneal Wound

After intraperitoneal and topical anesthesia, corneal lesions were performed on both eyes of 40 mice by laser induced photorefractive keratectomy (PRK) []. To induce the epithelial and stromal laser lesion, each eye was placed under a Wavelight Allegretto Wave PR-020407 excimer laser (Wavelight GmbH, Alcon, Erlangen, Germany). The PRK ablation parameters included a diameter of 2 mm to the central optic zone and a total depth of 45 µm to induce a uniform lesion affecting both epithelium and stroma. The laser began nasally and progressed temporally.

2.5. Treatment Regimen

All animals were treated with topical eye drops of azythromycin 1.5% (Azyter, Laboratoires Thea, Clemrmont-Ferrand, France) for antimicrobial prophylaxis two times daily for three days after lesion []. Eighty eyes of 40 mice were divided in four treatment groups (n = 20 eyes per group), which included control, stem cells, basic serum, and plasma rich in growth factor (PGRF). Data from the PGRF (not shown) were intended and collected for a different ongoing study. Control eyes received only antibiotic eye drops. The other 3 groups also received topical treatment applied three times a day for five consecutive days. Topical drops were administered with a delay of at least 5 min between applications for multiple treatment regimens. Stem cell topical eye drops were prepared daily with 1 × 105 cells suspended in 25 µL HBSS/treatment []. The basic serum group received topical application of 25 µL of 100% human serum.

2.6. Ocular Surface Evaluation

Upon topical anesthesia, each treated eye was examined with a stereo biomicroscope before application of topical treatment at 30, 54, 78, 100, and 172 h after lesion (also referred to in this study as day 1, 2, 3, 4, and 7, respectively). This was done at each time point to assess corneal inflammation, opacities, and other anterior surface complications (i.e., infection, perforation, etc.). Fluorescein sodium solution (Colircusí Fluoresceína, Alcon Cusí, Barcelona, Spain) was used to evaluate the degree of the corneal epithelial defect. Each animal’s anterior segment was photographed with a Leica S6D stereo microscope (6.3:1 zoom and 15.0× magnification) equipped with a Leica EC3 digital camera (Leica Microsystems, Wetzlar, Germany) with and without fluorescein at each clinical assessment. The defect area was determined by the fluorescein positive remaining area under blue light (1 mm = 240 pixels) using ImageJ 1.45a software (National Institutes of Health, Bethesda, MD, USA). Based on anterior segment visualization of pupil, iris and the presence of corneal vessels with stereo biomicroscopy at each examination time point. The analysis was performed independently by two masked graders.

2.7. Histological Examination

Eyes were fixed in Somogyi’s fixative without glutaraldehyde, rapidly frozen in liquid nitrogen, and preserved in OCT compound. The specimens were cut into 5 µm-thick tissue sections with a cryostat and subjected to immunofluorescence techniques. Sections were examined under fluorescence microscopy. With regards to the 33 mice of the 40 animals (66 eyes) included in the analysis, 32 eyes of 16 animals were enucleated after 78 h (day 3), while the remaining 34 eyes of 17 mice were enucleated at the end of the study at 172 h (day 7) post-lesion. The details regarding the histological preparation and assessment are reported in the Appendix section.
For immunofluorescence analysis we used antibodies to Ki67 (proliferation; 1:500; Abcam, Cambridge, UK), α-SMA (myofibroblast transformation; 1:200; Abcam), E-cadherin (assembly of epithelial cells; 1:200; Santa Cruz Biotechnology, Santa Cruz, CA, USA). Further details regarding the antibodies used in our study have been reported in the Appendix section under the heading “Histological examination” on page 14, lines 532–575.

3. Statistical Analysis

Normality of the data distribution was assessed with the Kolmogorov-Smirnov test. Data were expressed as median ± standard deviation. Differences of the data amongst groups were analyzed with SPSS 20.0 (SPSS Inc., Chicago, IL, USA) for Windows program using Kruskal-Wallis and Friedman test. Multiple comparisons were performed with Dunnett’s test. A p value of < 0.05 was considered to be statistically significant.

4. Results

4.1. Clinical Outcomes

The PRK lesion was performed uneventfully in all mice eyes. Immediately after the laser ablation, the treated areas showed a whitish uniform, hazy appearance. There were no signs of neovascularization or perforation in all eyes. Three animals (#24, 29 & 39) died of hypothermia at 30 h after anesthesia, and four animals (#6, 12, 38 & 28) were found dead in the cages (probably due to natural causes) before the endpoint on the 7th day. A total of 33 of 40 animals (66 eyes) were considered in the statistical analysis: 18 stem cell treated eyes, 15 basic serum eyes, and 17 control eyes. Of these, 16 (32 eyes) underwent treatment for three days and sacrificed at 78 h, and the remaining 17 animals (34 eyes) completed the five-day treatment regimen and then sacrificed at 172 h.
Partial re-epithelization was seen in all mice eyes at the first time point at 30 h. All eyes were completely re-epithelized by 100 h (Table 1a & Figure 1). On the first day, the fluorescein positive corneal lesion area was significantly smaller in the stem cells groups than the control eyes (Table 1a & Figure 1 & Figure 2p< 0.05); on the second day, it was significantly larger in the controls, yet comparable between stem cell and serum treatment groups (Table 1a & Figure 1 & Figure 2p < 0.02). No differences were found amongst groups on the other days.
Figure 1

Intragroup comparisons of median epithelium defect area over time for each treatment group in the mice eyes. * = statistically significant difference between stem cells and control groups; ^ = statistically significant difference between controls and 
Figure 2

Box plots of median epithelium defect area over time at each time point for each treatment group in the mice eyes. * = statistically significant difference between stem cells and control groups; ^ = statistically significant difference between controls 
Inter-individual differences were seen in the mice, even amongst eyes with the same treatment. To limit this variability, several mice were treated with stem cells on the right eye and control on the left. The stem cell treated eyes tended to show faster wound healing with smaller defect areas at most of the earlier time points in all these eyes. Figure 3 shows examples of the fluorescein positive areas at each time point for these eyes.
Figure 3

Fluorescein positive areas at each time for animals # 1, 2, 3, and 17. For intraindividual comparisons, the right eye (red squares) of each mouse was treated with stem cells and the left eye (blue triangles) was treated with the control regimen. The stem 
Qualitative histology assessments were performed in 27 eyes of 15 mice eyes at 78 h (day 3) after lesion (10 control, 8 stem, 9 basic serum) and 28 eyes of 15 mice eyes at 172 h (day 7) after lesion (8 control, 10 stem, 10 basic serum).

4.2. Histological Outcomes

With regards to the epithelium histological assessment after three days, the stem cell group showed a slightly increased number of epithelial cell layers (Figure 4 and Figure 5). There were three layers of epithelial cells in the peripheral region of the damaged cornea, while about four layers of cells in the central post-injured area. The number of Ki67 cells in the peripheral epithelium was similar amongst groups, however, the stem cells treated eyes showed less of these actively replicating cells in the epithelium, yet more in the stroma underlying the lesion when compared with the other groups (Table 1b). E-Cadherin protein accumulated in the basal cell layer of the epithelium and was present in the cytoplasm (Figure 5). This localization suggests that the epithelium was not stabilized and was still involved in a reorganization process [,,]. In the control eyes, there were only three layers of epithelial cells and a slightly less number of Ki67+ cells in the peripheral epithelium (Table 1b). The E-Cadherin pattern was similar to that described in the stem cell treated eyes (Figure 5). In the basic serum eyes, there were 5 to 6 layers of epithelial cells and a higher number of Ki67+ cells across the extension of the epithelium. The E-Cadherin pattern was diffuse in the basal cells of the epithelium, similar to the stain observed in the other groups.
Figure 4

Images of Ki67 positive cells (in red) in the central epithelium of wound healing corneas treated with adipose-derived stem cells ADSC for three days (A) compared to the control eyes (B). Both groups showed a high number of proliferating cells in the 
Figure 5

Corneal epithelium wound healing after three days assessed with E-Cadherin immunostaining. (A) represents an eye treated with ADSC. E-Cadherin (marked in red) appeared to be distributed in the basal cell layer of the epithelium and accumulated in the 
With regards to the stromal histology at three days, all eyes showed E-Cadherin-positive cells indicating the presence of migratory elements in the stroma, probably involved in reparative processes. Moreover, a small number of myofibroblasts (α-SMA positive profiles) were found in the stromal under the epithelium in the lesion area. These myofibroblasts were more frequent in the control corneas when compared to those found in the basic serum treated eyes. These myofibroblasts were scarce and not frequently seen in the stem cell treated eyes (Figure 6a).
Figure 6

(a) ADSC treatment (A) seemed to retard the onset of myofibroblast appearance when compared to the controls eyes (B) during the initial stages of the wound healing process (three days after injury). There tended to be scarce or no myofibroblasts in the 
After seven days of lesion, the stem cell treated eyes showed epithelium that was similar to uninjured epithelium, composed of 4 to 5 layers of uniform and perfectly structured epithelial cells (Figure 6b). E-Cadherin protein was localized at the periphery of epithelial cells, similar to what is normally found in healthy uninjured epithelium, thus indicative of a stabilization of the corneal regenerating epithelium []. The number and distribution of proliferating Ki67 positive cells were similar to that measured in normal mouse corneal epithelium. The control eyes showed the same structure of epithelium; however, there appeared to be at least one additional layer and more Ki67 positive cells (Table 1bFigure 7). The basic serum treated eyes showed about 5 to 6 layers in the epithelium and a higher number of Ki67 positive cells (Table 1b).
Figure 7

Left panel shows examples of proliferative events in the central epithelium 7 days after injury. In the ADSC treated eyes (A), there were scarce residual amounts of Ki67+ cells (in red, arrows), which can normally found in the epithelium uninjured corneas. 
The stromal histology after seven days (Figure 7) showed that the stem cell treated had little or no E-Cadherin positive elements in the stroma. The E-Cadherin positive elements, however, appeared to be abundant in the basic serum treated eyes and control eyes, which can be indicative of migratory events in the stroma. After seven days, there was a significant reduction in the quantity of α-SMA+ myofibroblasts in stem cell treated group (4.09 ± 0.48 cells) compared to control (10.85 ± 0.76 cells; p < 0.001) and, to a lesser extent, compared to basic serum group (6.27 ± 0.65 cells; p < 0.05).

5. Discussion

Similar to our previous studies, ADSC were obtained from human adipose tissue aspirates following a protocol optimized for the isolation and in vitro expansion of human multipotent adult stem cells [,]. ADSC expressed the pluripotent state-specific transcription factors Oct-4, Nanog and Sox 2 [,]. ADSC highly expressed CD90, CD105, CD73, however, were mainly negative for the hematopoietic markers CD34 and CD45. These cells displayed multipotency. Further details regarding ADSC are reported in the Appendix section. Our results show that ADSC seem to enhance corneal wound healing induced by laser ablation when compared to traditional topical therapy.
In accordance to our previous study based on chemical induced corneal lesions in rats [], the percent of epithelium fluorescein positive damage in eyes treated with stem cells (with or without serum) was smaller at each time point, which was statistically significant when compared to the control mice eyes (Table 1a). Stem cell treated eyes reached complete epithelium closure faster than the control eyes (Figure 1). The results showed statistically smaller lesions after day 1 in the stem cell group, which was comparable to the basic serum treated eyes but less than that observed in the control group on days 2 and 3 (Table 1aFigure 1 & Figure 2).
With regards to the mice histological data, the presence of more layers of epithelial cells in the early stages of repair in the stem cell treated group compared to the control eyes could be related to an improvement in the re-epithelization of the corneal surface after injury. The epithelium under the lesion had a smaller number of Ki67 actively replicating cells in the stem cell eyes, yet the stroma appeared to have significantly more than the other groups (Table 1b), which may be indicative of earlier stromal repair and post-mitotic epithelium layer reformation in this group at three days. Although these epithelial layers covered the entire corneal surface, these cells appeared to be undergoing reorganization during the initial period after lesion, as deduced by the elevated rate of proliferation of basal epithelial cells in the central region of the cornea and the localization pattern of E-Cadherin protein in the epithelium [,]. E-Cadherin did not appear to be distributed in the periphery of epithelial cells, as expected in stable uninjured epithelium; however, it mostly accumulated in the cytoplasm of basal epithelial cells. This location of E-Cadherin suggested an ongoing reorganization process of the epithelial layers [,,].
Although all eyes exhibited complete re-epithelization macroscopically after seven days, the histological evaluation showed that the epithelium of individuals treated with stem cell contained four or five layers, which is the number of layers normally found in uninjured mouse corneal epithelium. The number of dividing cells (Ki67+) in the central region was very low, which resembled a normal cornea with a well-established epithelium structure. The cytoarchitecture of the epithelium was that of a normal epithelium, showing cuboidal cells in the basal layer, with a few layers of low cuboidal cells positioned on top, followed by 1–2 outer sheets of flattened wing cells. E-Cadherin appeared to be located externally bordering epithelial cells and was observed in all layers. In comparison, the control eyes had at least one additional layer of epithelial cells, which may be indicative of an incomplete or altered repair process. The eyes treated with the blood derivatives PRGF (data not shown) and basic serum showed five to six layers of epithelial cells, which may be indicative of an ongoing re-epithelization and possible delay in the cessation of cell proliferation in the central region of the cornea, as seen with the presence of numerous Ki67+ cells.
The stroma of mice eyes treated with stem cells had a notably lower density of myofibroblasts (α-SMA+elements) compared with control and basic serum eyes at both three and seven days, which may be related to the slightly better corneal transparency and lower haze in the stem cell eyes (based on a qualitative comparative analysis between groups, data not shown) [,,,,]. E-Cadherin+ cells were not found in the stroma of stem cell treated eyes and controls after seven days, yet were abundant in the PRGF and serum treated eyes. The lack of E-Cadherin+ cells can be indicative of a stabilization of the stromal cytoarchitectural organization. In contrast, the stroma of the blood derived treated groups continued to have reorganizations events and mobility of cellular elements in the stroma. The physiological roles of ADSC are diverse and promising in tissue regeneration and wound healing [,,]. ADSC obtained from lipoaspirate have been shown to meet the minimal set of 4 criteria proposed by the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular therapy [] used to define functional human MSC [,,,] In addition to their extensive proliferation potential and multilineage differentiation, ADSC can interact and affect the immune system response to injury, by the down-regulation of proinflammatory factors and production of several trophic factors [,,]. Several reports have shown the immunoregulatory properties of MSC, which include the inhibition of T-cells [], increase in tumor necrosis factor (TNF) from dentritic cells [], increase in regulatory T-cells [], block of antigen producing cell maturation [], increase in immunosuppressive cytokine interleukin (IL)-10 and TNF-β and decrease in IL-2 []. The endocrine function of adipose tissue is evident through the secretion of numerous growth factors (GF) like epidermal GF, vascular endothelial GF, basic fibroblast GF, keratinocyte GF, and platelet-derived GF [].
There are limited studies in current literature regarding the use of ADSC in ocular surface and stromal wound healing [,,,]. Arnalich-Montiel showed that human ADSC could regenerate into corneal tissue in situ when inserted in a laser induced intrastromal corneal pocket in a rabbit model []. The transplanted cells appeared to be safe, maintained corneal transparency and caused no immune reaction []. The transplanted multipotent stem cells acquired similar characteristics to keratocytes []. Studies have shown that topical application of stem cells are easy, and may prove to be clinically acceptable and effective in acute phases [,].
The presence of corneal limbal stem cells (LSC) was first discovered in the late 1980’s []. These slow-cycling subpopulation of epithelial basal cells located in the peripheral limbus of the cornea were found to have a substantial proliferating capacity. Autologous and allogenic LSC transplants are surgical option [], however, donor tissues are limited and autologous transplants may give rise to iatrogenic damage to the healthy fellow eye or at times the fellow eye is not healthy. Recent studies have reported the presence of MSC in the human limbal biopsies having similar immunophenotype and immunocytochemical markers to those present in bone marrow derived MSC [].
With regards to the clinical use of ADSC for ocular surface wounds in humans, only a single case report has been published by a group in Greece []. A young male underwent an experimental treatment involving topical application of autologous ADSC, which was obtained by lipoaspiration of subcutaneous adipose tissue from the lumbar area. The ADSC were isolated from the lipoaspirate and applied to the bottom of the ulcer, followed by closure of the lid with a pressure eye patch for 24 h. Corneal healing was observed after 11 days. At six months after treatment, the patient still did not require surgery, visual acuity was improved to 20/40, central corneal thickness was 476 µm, and corneal transparency improved with mild residual anterior stromal opacification.

6. Conclusions

Our mice experiments based on a laser-induced lesion confirmed that stem cell treated eyes had significantly smaller epithelial defects at each time point, with better and faster re-epithelization and less inflammatory response compared to other treatment arms, which was already reported in our preliminary rat study based on a chemical induced lesion model. Literature in this field is limiting, thus additional studies using a different animal model and different type of lesion was proposed in this study to confirm preliminary experiments and report additional data. Our mice experiments were based on a larger group of animals assessed for a longer follow-up compared to our preliminary rat studies. Our study adds to the limiting literature currently available in this field by confirming the biosafety, immunogenicity, and potential clinical and histological advantages in using this mode of stem cell treatment. Future studies involve the assessment of epithelial recovery, inflammation, corneal haze, and quantitative histological assessments compared in different treatment arms. Although the exact mechanisms are not known, these cells are multipotent and have the potential to differentiate toward a keratocyte stromal lineage and therefore theoretically appear to be a promising therapeutic alternative and advantageous compared to treatments currently utilized in clinical practice. Most studies have shown ADSC to be promising in animal models, which can serve as a great stepping stone in addressing the topical therapeutic use of stem cells in humans.

Table 1

(a) Median fluorescein positive corneal lesion area (mm2) in mice for each group over time. (b) Number Ki67 cells in the peripheral and central post-lesion corneal epithelium and stroma in the different treatment groups at day 3 and day 7 after lesion 

Acknowledgments

We are very grateful for following people that collaborated in the study: Emanuele Rampino Cordaro, Fabrizio De Biasio and Lara Lazzaro for the assistance with obtaining the lipoaspirate tissue, Daniele Nigris for information regarding the hospital protocol and preparation of the human blood serum, and Annagrazia Adornetto, Federica Cavaliere, and Giuseppe Pasquale Varano for the assistance with the laboratory and animal experiments. Special thanks to the Fundación Ramón Areces. We would like to thank Thea Farma Spa, Milan, Italy for the assistance and collaboration.

Appendix A

Appendix A.1. Animals

Our decision to use both eyes was not solely based on a cost issue but based on a similar experimental model used in our previous study published in Current Eye Research []. This was done in accordance to the 3R statement of the European Community (reduce, replace, refine) that encourages the use of fewer experimental animals, and to compare treatments on the same animals to reduce inter-animal variability, thus both eyes were also used in the experiment. In addition, we obtained the approval of the Animal Ethical Committee of the University of Oviedo (Spain).

Appendix A.2. Histological Examination

Eyes were fixed in Somogyi’s fixative without glutaraldehyde for 3 h, cryoprotected overnight in 30% sucrose solution, embedded in OCT compound and frozen in liquid Nitrogen. The entire frozen eyeball was placed on a cryostat chunk and oriented to obtain transversal sections of the cornea. Cryostat sections with a thickness of 5 µm were collected on Superfrost microscope slides (Thermo Scientific, Rochester, NY, USA) and stored frozen.
For each eye, three selected histological sections in the central region of the injured cornea were chosen to demonstrate each marker used to evaluate the quality of corneal repair, which included:
  1. Polyclonal antibody against Ki67 produced in rabbit (Abcam, Cambridge, UK): This antibody is present in the nuclei of actively dividing cells and can be used as a measure of the number of proliferating cells in a repairing process [].
  2. Polyclonal antibody against α Smooth Muscle Actin (α-SMA) produced in rabbit (Abcam, Cambridge, UK): The α-SMA presenting cells are identified as myofibroblast during the wound healing process [,,].
  3. Polyclonal antibody against E-Cadherin produced in rabbit (Santa Cruz Biotechnology, Santa Cruz, CA, USA): The E-Cadherin protein locates normally bordering epithelial cells, thus allowing a visualization of the cytoarchitecture of a multilayered epithelium [,].
Fluorescent immunohistochemical techniques were used to assess these markers. Selected cryosections were carefully thawed in a water bath; slides were placed in a humidity chamber and washed three times in PBS (Phosphate Buffered Saline) with Triton X-100 (0.03%). A brief incubation with a blocking medium (1% bovine serum albumin and 10% normal goat serum; Vector Laboratories, Burlingame, CA, USA) was added to avoid nonspecific binding of the secondary antibody. The blocking medium was then removed by washing the slides three times with PBS with Triton X-100 (0.03%). The sections were incubated overnight at 4 °C with the appropriate primary antibody at a specific dilution (Anti-Ki67 at 1:500; Anti-αSMA at 1:200; and, Anti-E-Cadherin at 1:100). Sections were then washed again three times in PBS and incubated with a fluorescent secondary antibody (anti-rabbit Alexa Fluor 594 raised in goat, Molecular Probes, Grand Island, NY, USA) in a dilution of 1:500 for 2 h at room temperature in the dark. Three additional washes were then used to eliminate the excess of fluorescent antibody. Nuclei were counterstained with 2 µg/mL 4′,6-diamidino-2-phenylindole dihydrochloride (DAPI; Molecular Probes, Grand Island, NY, USA) for 10 min and slides were mounted with Dako fluorescence mounting medium (Dako, Glostrup, Denmark) and coverslips. Slides were then identified and encoded with a numerical system and stored at 4 °C.
The analysis of the wound healing parameters was performed on histological images using a Leica DM6000 microscope equipped with epifluorescence using a 20× objective (numerical aperture 0.45) and AF6000 software (Leica Microsystems, Wetzlar, Germany). Measurements were performed in a semiautomated manner using ImageJ 1.45a software (National Institutes of Health, Bethesda, MD, USA). Two independent masked observers performed the analysis. Data were collected and decoded, and statistical analysis was performed using InStat 3.1a software (GraphPad Software Inc., La Jolla, CA, USA).

Appendix A.3. Adipose-Derived Stem Cells (ADSC)

Human subcutaneous abdominal adipose tissue was obtained from thee healthy female patients (aged 35, 52, and 64 years) undergoing elective lipoaspiration surgery. ADSC used in the treatment group, however, came from a single healthy donor (youngest female aged 35 years) and were employed, at the third passage in culture, in their undifferentiated state. Cells were characterized in accordance to the study reported by Domenis et al. in 2015 []. Specifically, cells satisfied the minimal criteria of ISCT. Moreover, they were checked for additional markers, such as the expression of pluripotent state-specific transcription factors. Although we are aware that a certain degree of controversy surrounds this latter feature, Chih-Chien Tsai et al. [] has shown that Oct-4 is not only expressed but also required for MSC self-renewal. Additionally, other independent groups showed the expression of these markers in MSC [,].

Author Contributions

All coauthors contributed in all aspects of the study and in the preparation of the manuscript. The manuscript is based on a Ph.D. thesis by the principle author, Marco Zeppieri, which was deposited and rendered available on the University “open UniUd portal.” It has not been published in any other journal.

Conflicts of Interest

The authors declare no conflict of interest.

References

1. De Miguel M.P., Alio J.L., Arnalich-Montiel F., Fuentes-Julian S., de Benito-Llopis L., Amparo F., Bataille L. Cornea and ocular surface treatment. Curr. Stem Cell Res. Ther. 2010;5:195–204. doi: 10.2174/157488810791268663. [PubMed] [Cross Ref]
2. Oh J.Y., Kim M.K., Shin M.S., Lee H.J., Ko J.H., Wee W.R., Lee J.H. The anti-inflammatory and anti-angiogenic role of mesenchymal stem cells in corneal wound healing following chemical injury. Stem Cells. 2008;26:1047–1055. doi: 10.1634/stemcells.2007-0737. [PubMed] [Cross Ref]
3. Ma Y., Xu Y., Xiao Z., Yang W., Zhang C., Song E., Du Y., Li L. Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. Stem Cells. 2006;24:315–321. doi: 10.1634/stemcells.2005-0046. [PubMed] [Cross Ref]
4. Geerling G., Maclennan S., Hartwig D. Autologous serum eye drops for ocular surface disorders. Br. J. Ophthalmol. 2004;88:1467–1474. doi: 10.1136/bjo.2004.044347. [PMC free article] [PubMed] [Cross Ref]
5. Anitua E., Sanchez M., Merayo-Lloves J., De la Fuente M., Muruzabal F., Orive G. Plasma rich in growth factors (PRGF-Endoret) stimulates proliferation and migration of primary keratocytes and conjunctival fibroblasts and inhibits and reverts TGF-beta1-Induced myodifferentiation. Investig. Ophthalmol. Vis. Sci. 2011;52:6066–6073. doi: 10.1167/iovs.11-7302. [PubMed] [Cross Ref]
6. Tanidir S.T., Yuksel N., Altintas O., Yildiz D.K., Sener E., Caglar Y. The effect of subconjunctival platelet-rich plasma on corneal epithelial wound healing. Cornea. 2010;29:664–669. doi: 10.1097/ICO.0b013e3181c29633. [PubMed] [Cross Ref]
7. Reimondez-Troitiño S., Alcalde I., Csaba N., Íñigo-Portugués A., de la Fuente M., Bech F., Riestra A.C., Merayo-Lloves J., Alonso M.J. Polymeric nanocapsules: A potential new therapy for corneal wound healing. Drug Deliv. Transl. Res. 2016;6:708–721. doi: 10.1007/s13346-016-0312-0. [PubMed][Cross Ref]
8. Alcalde I., Íñigo-Portugués A., Carreño N., Riestra A.C., Merayo-Lloves J.M. Effects of new biomimetic regenerating agents on corneal wound healing in an experimental model of post-surgical corneal ulcers. Arch. Soc. Esp. Oftalmol. 2015;90:467–474. doi: 10.1016/j.oftal.2015.04.006. [PubMed][Cross Ref]
9. Casteilla L., Planat-Benard V., Laharrague P., Cousin B. Adipose-derived stromal cells: Their identity and uses in clinical trials, an update. World J. Stem Cells. 2011;3:25–33. doi: 10.4252/wjsc.v3.i4.25.[PMC free article] [PubMed] [Cross Ref]
10. Gimble J.M., Guilak F., Bunnell B.A. Clinical and preclinical translation of cell-based therapies using adipose tissue-derived cells. Stem Cell Res. Ther. 2010;1:19. doi: 10.1186/scrt19. [PMC free article][PubMed] [Cross Ref]
11. Philippe B., Luc S., Valerie P.B., Jérôme R., Alessandra B.R., Louis C. Culture and Use of Mesenchymal Stromal Cells in Phase I and II Clinical Trials. Stem Cells Int. 2010;2010:503593. doi: 10.4061/2010/503593. [PMC free article] [PubMed] [Cross Ref]
12. Beltrami A.P., Cesselli D., Bergamin N., Marcon P., Rigo S., Puppato E., D’Aurizio F., Verardo R., Piazza S., Pignatelli A., et al. Multipotent cells can be generated in vitro from several adult human organs (heart, liver, and bone marrow) Blood. 2007;110:3438–3446. doi: 10.1182/blood-2006-11-055566.[PubMed] [Cross Ref]
13. Zuk P.A., Zhu M., Ashjian P., De Ugarte D.A., Huang J.I., Mizuno H., Alfonso Z.C., Fraser J.K., Benhaim P., Hedrick M.H. Human adipose tissue is a source of multipotent stem cells. Mol. Biol. Cell. 2002;13:4279–4295. doi: 10.1091/mbc.E02-02-0105. [PMC free article] [PubMed] [Cross Ref]
14. Zuk P.A. The adipose-derived stem cell: Looking back and looking ahead. Mol. Biol. Cell. 2010;21:1783–1787. doi: 10.1091/mbc.E09-07-0589. [PMC free article] [PubMed] [Cross Ref]
15. Sakaguchi Y., Sekiya I., Yagishita K., Muneta T. Comparison of human stem cells derived from various mesenchymal tissues: Superiority of synovium as a cell source. Arthritis Rheumatol. 2005;52:2521–2529. doi: 10.1002/art.21212. [PubMed] [Cross Ref]
16. Arnalich-Montiel F., Pastor S., Blazquez-Martinez A., Fernandez-Delgado J., Nistal M., Alio J.L., De Miguel M.P. Adipose-derived stem cells are a source for cell therapy of the corneal stroma. Stem Cells. 2008;26:570–579. doi: 10.1634/stemcells.2007-0653. [PubMed] [Cross Ref]
17. Zeppieri M., Salvetat M.L., Beltrami A.P., Cesselli D., Bergamin N., Russo R., Cavaliere F., Varano G.P., Alcalde I., Merayo J., et al. Human adipose-derived stem cells for the treatment of chemically burned rat cornea: Preliminary results. Curr. Eye Res. 2013;38:451–463. doi: 10.3109/02713683.2012.763100.[PubMed] [Cross Ref]
18. Ferro F., Spelat R., Falini G., Gallelli A., D’Aurizio F., Puppato E., Pandolfi M., Beltrami A.P., Cesselli D., Beltrami C.A., Ambesi-Impiombato F.S. Adipose tissue-derived stem cell in vitro differentiation in a three-dimensional dental bud structure. Am. J. Pathol. 2011;178:2299–2310. doi: 10.1016/j.ajpath.2011.01.055. [PMC free article] [PubMed] [Cross Ref]
19. Merayo-Lloves J., Blanco-Mezquita T., Ibares-Frias L., Cantalapiedra-Rodríguez R., Alvarez-Barcia A. Efficacy and safety of short-duration topical treatment with azithromycin oil-based eyedrops in an experimental model of corneal refractive surgery. Eur. J. Ophthalmol. 2010;20:979–988. [PubMed]
20. Li L., Hartley R., Reiss B., Sun Y., Pu J., Wu D., Lin F., Hoang T., Yamada S., Jiang J., et al. E-cadherin plays an essential role in collective directional migration of large epithelial sheets. Cell. Mol. Life Sci. 2012;69:2779–2789. doi: 10.1007/s00018-012-0951-3. [PMC free article] [PubMed] [Cross Ref]
21. Tian X., Liu Z., Niu B., Zhang J., Tan T.K., Lee S.R., Zhao Y., Harris D.C., Zheng G. E-cadherin/beta-catenin complex and the epithelial barrier. J. Biomed. Biotechnol. 2011;2011:567305. doi: 10.1155/2011/567305. [PMC free article] [PubMed] [Cross Ref]
22. Baum B., Georgiou M. Dynamics of adherens junctions in epithelial establishment, maintenance, and remodeling. J. Cell Biol. 2011;192:907–917. doi: 10.1083/jcb.201009141. [PMC free article] [PubMed][Cross Ref]
23. Du Y., Carlson E.C., Funderburgh M.L., Birk D.E., Pearlman E., Guo N. Stem cell therapy restores transparency to defective murine corneas. Stem Cells. 2009;27:1635–1642. doi: 10.1002/stem.91.[PMC free article] [PubMed] [Cross Ref]
24. Martinez-Garcia M.C., Merayo-Lloves J., Blanco-Mezquita T., Mar-Sardaña S. Wound healing following refractive surgery in hens. Exp. Eye Res. 2006;83:728–735. doi: 10.1016/j.exer.2006.02.017.[PubMed] [Cross Ref]
25. Jester J.V., Petroll W.M., Cavanagh H.D. Corneal stromal wound healing in refractive surgery: The role of myofibroblasts. Prog. Retin. Eye Res. 1999;18:311–356. doi: 10.1016/S1350-9462(98)00021-4.[PubMed] [Cross Ref]
26. Hassell J.R., Birk D.E. The molecular basis of corneal transparency. Exp. Eye Res. 2010;91:326–335. doi: 10.1016/j.exer.2010.06.021. [PMC free article] [PubMed] [Cross Ref]
27. Merayo-Lloves J., Yanez B., Mayo A., Martín R., Pastor J.C. Experimental model of corneal haze in chickens. J. Refract. Surg. 2001;17:696–699. [PubMed]
28. Domenis R., Lazzaro L., Calabrese S., Mangoni D., Gallelli A., Bourkoula E., Manini I., Bergamin N., Toffoletto B., Beltrami C.A., et al. Adipose tissue derived stem cells: In vitro and in vivo analysis of a standard and three commercially available cell-assisted lipotransfer techniques. Stem Cell Res. Ther. 2015;6:2–15. doi: 10.1186/scrt536. [PMC free article] [PubMed] [Cross Ref]
29. Dominici M.L., Le Blanc K., Mueller I., Slaper-Cortenbach I., Marini F.C., Krause D.S., Deans R.J., Keating A., Prockop D.J., Horwitz E.M. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–317. doi: 10.1080/14653240600855905. [PubMed] [Cross Ref]
30. Tobita M., Orbay H., Mizuno H. Adipose-derived stem cells: Current findings and future perspectives. Discov. Med. 2011;11:160–170. [PubMed]
31. Meirelles L.S., Fontes A.M., Covas D.T., Caplan A.I. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev. 2009;20:419–427. doi: 10.1016/j.cytogfr.2009.10.002. [PubMed] [Cross Ref]
32. Bunnell B.A., Flaat M., Gagliardi C., Patel B., Ripoll C. Adipose-derived stem cells: Isolation, expansion and differentiation. Methods. 2008;45:115–120. doi: 10.1016/j.ymeth.2008.03.006.[PMC free article] [PubMed] [Cross Ref]
33. Di Nicola M., Carlo-Stella C., Magni M., Milanesi M., Longoni P.D., Matteucci P., Grisanti S., Gianni A.M. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838–3843. doi: 10.1182/blood.V99.10.3838. [PubMed][Cross Ref]
34. Aggarwal S., Pittenger M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–1822. doi: 10.1182/blood-2004-04-1559. [PubMed] [Cross Ref]
35. Zhang W., Ge W., Li C., You S., Liao L., Han Q., Deng W., Zhao R.C. endritic cells. Stem Cells Dev. 2004;13:263–271. doi: 10.1089/154732804323099190. [PubMed] [Cross Ref]
36. Harkin D.G., Foyn L., Bray L.J., Sutherland A.J., Li F.J., Cronin B.G. Concise reviews: Can mesenchymal stromal cells differentiate into corneal cells? A systematic review of published data. Stem Cells. 2015;33:785–791. doi: 10.1002/stem.1895. [PubMed] [Cross Ref]
37. Ghieh F., Jurjus R., Ibrahim A., Geagea A.G., Daouk H., El Baba B., Chams S., Matar M., Zein W., Jurjus A. The Use of Stem Cells in Burn Wound Healing: A Review. Biomed Res. Int. 2015 doi: 10.1155/2015/684084. [PMC free article] [PubMed] [Cross Ref]
38. Ljubimov A.V., Saghizadeh M. Progress in corneal wound healing. Prog. Retin. Eye Res. 2015;49:17–45. doi: 10.1016/j.preteyeres.2015.07.002. [PMC free article] [PubMed] [Cross Ref]
39. Bermudez M.A., Sendon-Lago J., Eiro N., Trevino M., Gonzalez F., Yebra-Pimentel E., Giraldez M.J., Macia M., Lamelas M.L., Saa J., et al. Corneal epithelial wound healing and bactericidal effect of conditioned medium from human uterine cervical stem cells. Investig. Ophthalmol. Vis. Sci. 2015;56:983–992. doi: 10.1167/iovs.14-15859. [PubMed] [Cross Ref]
40. Patel S.A., Sherman L., Munoz J., Rameshwar P. Immunological properties of mesenchymal stem cells and clinical implications. Arch. Immunol. Ther. Exp. (Warsz) 2008;56:1–8. doi: 10.1007/s00005-008-0001-x. [PubMed] [Cross Ref]
41. Boquest A.C., Shahdadfar A., Frønsdal K., Sigurjonsson O., Tunheim S.H., Collas P., Brinchmann J.E. Isolation and transcription profiling of purified uncultured human stromal stem cells: Alteration of gene expression after in vitro cell culture. Mol. Biol. Cell. 2005;16:1131–1141. doi: 10.1091/mbc.E04-10-0949.[PMC free article] [PubMed] [Cross Ref]
42. Du Y., Roh D.S., Funderburgh M.L., Mann M.M., Marra K.G., Rubin J.P. Adipose-derived stem cells differentiate to keratocytes in vitro. Mol. Vis. 2010;16:2680–2689. [PMC free article] [PubMed]
43. Acar U., Pinarli F.A., Acar D.E., Beyazyildiz E., Sobaci G., Ozgermen B.B. Effect of allogeneic limbal mesenchymal stem cell therapy in corneal healing: Role of administration route. Ophthalmic Res. 2015;53:82–89. doi: 10.1159/000368659. [PubMed] [Cross Ref]
44. Cotsarelis G., Cheng S.Z., Dong G., Sun T.T., Lavker R.M. Existence of slow-cycling limbal epithelial basal cells that can be preferentially stimulated to proliferate: Implications on epithelial stem cells. Cell. 1989;57:201–209. doi: 10.1016/0092-8674(89)90958-6. [PubMed] [Cross Ref]
45. Kenyon K.R., Tseng S.C. Limbal autograft transplantation for ocular surface disorders. Ophthalmology. 1989;96:709–722. [PubMed]
46. Polisetty N., Fatima A., Madhira S.L., Sangwan V.S., Vemuganti G.K. Mesenchymal cells from limbal stroma of human eye. Mol. Vis. 2008;14:431–442. [PMC free article] [PubMed]
47. Agorogiannis G.I., Alexaki V.I., Castana O., Kymionis G.D. Topical application of autologous adipose-derived mesenchymal stem cells (MSCs) for persistent sterile corneal epithelial defect. Graefes Arch. Clin. Exp. Ophthalmol. 2012;250:455–457. doi: 10.1007/s00417-011-1841-3. [PubMed] [Cross Ref]
48. Puccinelli T.J., Bertics P.J., Masters K.S. Regulation of keratinocyte signaling and function via changes in epidermal growth factor presentation. Acta Biomater. 2010;6:3415–3425. doi: 10.1016/j.actbio.2010.04.006. [PMC free article] [PubMed] [Cross Ref]
49. Myrna K.E., Pot S.A., Murphy C.J. Meet the corneal myofibroblast: The role of myofibroblast transformation in corneal wound healing and pathology. Vet. Ophthalmol. 2009;12(Suppl. 1):25–27. doi: 10.1111/j.1463-5224.2009.00742.x. [PMC free article] [PubMed] [Cross Ref]
50. Tsai C.C., Su P.F., Huang Y.F., Yew T.L., Hung S.C. Oct4 and Nanog directly regulate Dnmt1 to maintain self-renewal and undifferentiated state in mesenchymal stem cells. Mol. Cell. 2012;47:169–182. doi: 10.1016/j.molcel.2012.06.020. [PubMed] [Cross Ref]
51. Wei X., Shen C.Y. Transcriptional regulation of oct4 in human bone marrow mesenchymal stem cells. Stem Cells Dev. 2011;20:441–449. doi: 10.1089/scd.2010.0069. [PubMed] [Cross Ref]
52. Han S.M., Han S.H., Coh Y.R., Jang G., Chan Ra J., Kang S.K. Enhanced proliferation and differentiation of Oct4- and Sox2-overexpressing human adipose tissue mesenchymal stem cells. Exp. Mol. Med. 2014;46:e101. doi: 10.1038/emm.2014.28. [PMC free article] [PubMed] [Cross Ref]

2.

Graefe’s Archive for Clinical and Experimental OphthalmologyVolume 250, Issue 3, March 2012, Pages 455-457

Topical application of autologous adipose-derived mesenchymal stem cells (MSCs) for persistent sterile corneal epithelial defect(Article)

  • aEye Clinic, University Hospital of Heraklion, Crete, Heraklion, Greece
  • bLaboratory of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
  • cDepartment of Plastic and Reconstructive Surgery, Evangelismos General Hospital, Athens, Greece





PLOS: http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0186238

Human adipose-derived stem cells support the growth of limbal stem/progenitor cells

  • Published: October 11, 2017

Abstract

The most efficient method to expand limbal stem cells (LSCs) in vitro for clinical transplantation is to culture single LSCs directly on growth-arrested mouse fibroblast 3T3 cells. To reduce possible xenobiotic contamination from 3T3s, primary human adipose-derived stem cells (ASCs) were examined as feeder cells to support the expansion of LSCs in vitro. To optimize the ASC-supported culture, freshly isolated limbal epithelial cells in the form of single cells (SC-ASC) or cell clusters (CC-ASC) were cultured using three different methods: LSCs seeded directly on feeder cells, a 3-dimensional (3D) culture system and a 3D culture system with fibrin (fibrin 3D). The expanded LSCs were examined at the end of a 2-week culture. The standard 3T3 culture served as control. Expansion of SC-ASC showed limited proliferation and exhibited differentiated morphology. CC-ASC generated epithelial cells with undifferentiated morphology in all culture methods, among which CC-ASC in 3D culture supported the highest cell doubling (cells doubled 9.0 times compared to cells doubled 4.9 times in control) while maintained the percentage of putative limbal stem/progenitor cells compared to the control. There were few cell-cell contacts between cultured LSCs and ASCs in 3D CC-ASC. In conclusion, ASCs support the growth of LSCs in the form of cell clusters but not in single cells. 3D CC-ASC could serve as a substitute for the standard 3T3 culture to expand LSCs.
2.
 2018 Jan 9;27(1):68-83. doi: 10.1016/j.cmet.2017.12.002.

Anatomical, Physiological, and Functional Diversity of Adipose Tissue.

Author information

1
Department of Molecular, Cellular, and Developmental Biology, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA.
2
Department of Developmental and Cell Biology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.
3
Department of Molecular, Cellular, and Developmental Biology, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA; Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA. Electronic address: valerie.horsley@yale.edu.
4
Department of Developmental and Cell Biology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA. Electronic address: plikus@uci.edu.

Abstract

Adipose tissue depots can exist in close association with other organs, where they assume diverse, often non-traditional functions. In stemcell-rich skin, bone marrow, and mammary glands, adipocytes signal to and modulate organ regeneration and remodeling. Skin adipocytes and their progenitors signal to hair follicles, promoting epithelial stem cell quiescence and activation, respectively. Hair follicles signal back to adipocyte progenitors, inducing their expansion and regeneration, as in skin scars. In mammary glands and heart, adipocytes supply lipids to neighboring cells for nutritional and metabolic functions, respectively. Adipose depots adjacent to skeletal structures function to absorb mechanical shock. Adipose tissue near the surface of skin and intestine senses and responds to bacterial invasion, contributing to the body’s innate immune barrier. As the recognition of diverse adipose depot functions increases, novel therapeutic approaches centered on tissue-specific adipocytes are likely to emerge for a range of cancers and regenerative, infectious, and autoimmune disorders.

3.

 2017 Nov;5(6):406-418.

Effects of Human Adipose-derived Stem Cells and Platelet-Rich Plasma on Healing Response of Canine Alveolar Surgical Bone Defects.

Author information

1
Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.

Abstract

BACKGROUND:

Due to the known disadvantages of autologous bone grafting, tissue engineering approaches have become an attractive method for ridge augmentation in dentistry. To the best of our knowledge, this is the first study conducted to evaluate the potential therapeutic capacity of PRP-assisted hADSCs seeded on HA/TCP granules on regenerative healing response of canine alveolar surgical bone defects. This could offer a great advantage to alternative approaches of bone tissue healing-induced therapies at clinically chair-side procedures.

METHODS:

Cylindrical through-and-through defects were drilled in the mandibular plate of 5 mongrel dogs and filled randomly as following: I- autologous crushed mandibular bone, II- no filling material, III- HA/TCP granules in combination with PRP, and IV- PRP-enriched hADSCs seeded on HA/TCP granules. After the completion of an 8-week period of healing, radiographic, histological and histomorphometrical analysis of osteocyte number, newly-formed vessels and marrow spaces were used for evaluation and comparison of the mentioned groups. Furthermore, the buccal side of mandibular alveolar bone of every individual animal was drilled as normal control samples (n=5).

RESULTS:

Our results revealed that hADSCs subcultured on HA/TCP granules in combination with PRP significantly promoted bone tissue regeneration as compared with those defects treated only with PRP and HA/TCP granules (P<0.05).

CONCLUSION:

In conclusion, our results indicated that application of PRP-assisted hADSCs could induce bone tissue regeneration in canine alveolar bone defects and thus, present a helpful alternative in bone tissue regeneration.

 2017;2017:2156478. doi: 10.1155/2017/2156478. Epub 2017 Dec 14.

Impact of Tissue Harvesting Sites on the Cellular Behaviors of Adipose-Derived Stem Cells: Implication for Bone Tissue Engineering.

Author information

1
Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
2
Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
3
Department of Applied Cell Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.

Abstract

The advantages of adipose-derived stem cells (AdSCs) over bone marrow stem cells (BMSCs), such as being available as a medical waste and less discomfort during harvest, have made them a good alternative instead of BMSCs in tissue engineering. AdSCs from buccal fat pad (BFP), as an easily harvestable and accessible source, have gained interest to be used for bone regeneration in the maxillofacial region. Due to scarcity of data regarding comparative analysis of isolated AdSCs from different parts of the body, we aimed to quantitatively compare the proliferation and osteogenic capabilities of AdSCs from different harvesting sites. In this study, AdSCs were isolated from BFP (BFPdSCs), abdomen (abdomen-derived mesenchymal stem cells (AbdSCs)), and hip (hip-derived mesenchymal stem cells (HdSCs)) from one individual and were compared for surface marker expression, morphology, growth rate, and osteogenic differentiation capability. Among them, BFPdSCs demonstrated the highest proliferation rate with the shortest doubling time and also expressed vascular endothelial markers including CD34 and CD146. Moreover, the expression of osteogenic markers were significantly higher in BFPdSCs. The results of this study suggested that BFPdSCs as an encouraging source of mesenchymal stem cells are to be used for bone tissue engineering.

 2017 Dec;14(6):5956-5964. doi: 10.3892/etm.2017.5333. Epub 2017 Oct 18.

Comparison of bone marrow-vs. adipose tissue-derived mesenchymal stem cells for attenuating liver fibrosis.

Hao T1Chen J2Zhi S1Zhang Q1Chen G1Yu F1.

Author information

1
Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China.
2
Department of Anorectal Surgery, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 325000, P.R. China.

Abstract

Mesenchymal stem cell (MSC) therapy has emerged as a potential novel method of treating liver fibrosis. To date, bone marrow-derived MSCs (BM-MSCs) and adipose tissue-derived MSCs (AD-MSCs) have not been analyzed with respect to their ability to combat liver fibrosis. The present study aimed to compare the capabilities of BM-MSCs and AD-MSCs in the treatment of liver fibrosis. BM-MSCs and AD-MSCs were taken from male Sprague-Dawley rats and cultured. Hepatic stellate cells (HSCs) were co-cultured with either BM-MSCs or AD-MSCs, and the effects of BM-MSCs or AD-MSCs on the proliferation, activation and apoptosis of HSCs were determined. The secretion of a selected group of cytokines by BM-MSCs and AD-MSCs was measured using enzyme-linked immunosorbent assays. Using a CCl4-induced liver fibrosis animal model, the anti-inflammatory and anti-fibrotic effects of BM-MSCs or AD-MSCs against liver fibrosis in vivo were evaluated. The morphological examination and analysis of specific surface markers confirmed the successful preparation of BM-MSCs and AD-MSCs. Furthermore, the proliferation, activation and apoptosis of HSCs were significantly inhibited by BM-MSCs and AD-MSCs, with statistically greater reductions achieved by AD-MSCs compared with BM-MSCs. Direct comparison of the secretion of selected cytokines by BM-MSCs and AD-MSCs revealed that significantly higher levels of nerve growth factor and transforming growth factor-β1 were secreted in the AD-MSC culture medium, whereas levels of vascular endothelial growth factor and interleukin-10 did not differ significantly between AD-MSCs and BM-MSCs. In vivo studies using a CCl4-induced liver fibrosis model demonstrated that inflammatory activity and fibrosis staging scores were significantly lower in the MSC-treated groups compared with controls. Although AD-MSCs improved anti-inflammatory and anti-fibrotic effects compared with BM-MSCs, these differences were not significant. Thus, the current study demonstrated that BM-MSCs and AD-MSCs are similarly effective at attenuating liver fibrosis by inhibiting the activation and proliferation of HSCs, as well as promoting the apoptosis of HSCs.

 2017 Dec 1;21(2):122-127. doi: 10.5935/0946-5448.20170023.

Comparison of Three Types of Mesenchymal Stem Cells (Bone MarrowAdipose Tissue, and Umbilical Cord-Derived) as Potential Sources for Inner Ear Regeneration.

Author information

1
Department of Genetics and Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
2
Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.

Abstract

In this review, we compared the potential of mesenchymal stem cells derived from bone marrowadipose tissue and umbilical cord as suitable sources for regeneration of inner ear hair cells and auditory neurons. Our intensive literature search indicates that stem cells in some of adult mammalian tissues, such as bone marrow, can generate new cells under physiological and pathological conditions. Among various types of stem cellsbone marrow-derived mesenchymal stem cells are one of the most promising candidates for cell replacement therapy. Mesenchymal stem cells have been reported to invade the damaged area, contribute to the structural reorganization of the damaged cochlea and improve incomplete hearing recovery. We suggest that bone marrow-derived mesenchymal stem cells would be more beneficial than other mesenchymal stem cells.

 2018 Jan 14. pii: S8756-3282(18)30008-5. doi: 10.1016/j.bone.2018.01.008. [Epub ahead of print]

Development, regulation, metabolism and function of bone marrow adipose tissues.

Author information

1
Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States.
2
Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, MO, United States.
3
Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States. Electronic address: macdouga@umich.edu.

Abstract

Most adipocytes exist in discrete depots throughout the body, notably in well-defined white and brown adipose tissues. However, adipocytes also reside within specialized niches, of which the most abundant is within bone marrow. Whereas bone marrow adipose tissue (BMAT) shares many properties in common with white adipose tissue, the distinct functions of BMAT are reflected by its development, regulation, protein secretion, and lipid composition. In addition to its potential role as a local energy reservoir, BMAT also secretes proteins, including adiponectin, RANK ligand, dipeptidyl peptidase-4, and stem cell factor, which contribute to local marrow niche functions and which may also influence global metabolism. The characteristics of BMAT are also distinct depending on whether marrow adipocytes are contained within yellow or red marrow, as these can be thought of as ‘constitutive’ and ‘regulated’, respectively. The rBMAT for instance can be expanded or depleted by myriad factors, including age, nutrition, endocrine status and pharmaceuticals. Herein we review the site specificity, age-related development, metabolic characteristics and regulation of BMAT under various metabolic conditions, including the functional interactions with bone and hematopoietic cells.

Stem Cells Dev. 2012 Sep 20;21(14):2724-52. doi: 10.1089/scd.2011.0722. Epub 2012 May 9.

Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells.

Abstract

Mesenchymal stem/stromal cells (MSCs) comprise a heterogeneous population of cells with multilineage differentiation potential, the ability to modulate oxidative stress, and secrete various cytokines and growth factors that can have immunomodulatory, angiogenic, anti-inflammatory and anti-apoptotic effects. Recent data indicate that these paracrine factors may play a key role in MSC-mediated effects in modulating various acute and chronic pathological conditions. MSCs are found in virtually all organs of the body. Bone marrow-derived MSCs (BM-MSCs) were discovered first, and the bone marrow was considered the main source of MSCs for clinical application. Subsequently, MSCs have been isolated from various other sources with the adipose tissue, serving as one of the alternatives to bone marrow. Adipose tissue-derived MSCs (ASCs) can be more easily isolated; this approach is safer, and also, considerably larger amounts of ASCs can be obtained compared with the bone marrow. ASCs and BM-MSCs share many biological characteristics; however, there are some differences in their immunophenotype, differentiation potential, transcriptome, proteome, and immunomodulatory activity. Some of these differences may represent specific features of BM-MSCs and ASCs, while others are suggestive of the inherent heterogeneity of both BM-MSC and ASC populations. Still other differences may simply be related to different isolation and culture protocols. Most importantly, despite the minor differences between these MSC populations, ASCs seem to be as effective as BM-MSCs in clinical application, and, in some cases, may be better suited than BM-MSCs. In this review, we will examine in detail the ontology, biology, preclinical, and clinical application of BM-MSCs versus ASCs.


J Trauma Acute Care Surg. 2017 Apr 27. doi: 10.1097/TA.0000000000001489. [Epub ahead of print]

Differential inflammatory networks distinguish responses to bone marrow-derived vs. adipose-derived mesenchymal stem cell therapies in vascularized composite allotransplantation.

Abstract

BACKGROUND:

Vascularized composite allotransplantation (VCA) is aimed at enabling injured individuals to return to their previous lifestyles. Unfortunately, VCA induces an immune/inflammatory response, which mandates lifelong, systemic immunosuppression, with attendant detrimental effects. Mesenchymal stem cells (MSC) – both adipose-derived (AD-MSC) and bone marrow-derived (BM-MSC) – can reprogram inflammation and have been suggested as an alternative to immunosuppression, but their mechanism of action is as yet not fully elucidated. We sought to gain insights into these mechanisms using a systems biology approach.

METHODS:

PKH26 (red) dye-labeled AD-MSC or BM-MSC were administered intravenously to Lewis rat recipients of mismatched Brown Norway hindlimb transplants. Short course tacrolimus (FK-506) monotherapy was withdrawn at POD 21. Sera were collected at 4, 6, 18 weeks, assayed for 29 inflammatory/immune mediators, and the resultant data were analyzed using Dynamic Network Analysis (DyNA), Dynamic Bayesian Network (DyBN) inference, and Principal Component Analysis (PCA).

RESULTS:

DyNA network complexity decreased with time in AD-MSC rats, but increased in BM-MSC rats. DyBN and PCA suggested mostly different central nodes and principal characteristics, respectively, in AD-MSC vs. BM-MSC rats.

CONCLUSIONS:

AD-MSC and BM-MSC are associated with both overlapping and distinct dynamic networks and principal characteristics of inflammatory/immune mediators in VCA grafts with short course tacrolimus induction therapy. The decreasing inflammatory complexity of dynamic networks in the presence of AD-MSC supports the previously suggested role for regulatory T cells induced by AD-MSC. The finding of some overlapping and some distinct central nodes and principal characteristics suggests the role of key mediators in the response to VCA in general, as well as potentially differential roles for other mediators ascribed to the actions of the different MSC populations. Thus, combined in vivo/in silico strategies may yield novel means of optimizing MSC therapy for VCA


Which is the best method to obtain adipose derived stem cells?

 2015 Jan 5;6:2. doi: 10.1186/scrt536.

Adipose tissue derived stem cells: in vitro and in vivo analysis of a standard and three commercially available cell-assisted lipotransfer techniques.

Author information

1
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. rossana.domenis@uniud.it.
2
Clinic of Plastic and Reconstructive Surgery of Udine, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. laralazzaro@hotmail.com.
3
Clinic of Plastic and Reconstructive Surgery of Udine, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. sarah2012@hotmail.com.
4
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. damianomng@gmail.com.
5
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. byoteck@libero.it.
6
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. jenny_bourkoula@hotmail.com.
7
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. ivana.manini@tiscali.it.
8
Azienda Ospedaliero-Universitaria of Udine, P.le S. Maria della Misericordia 15, 33100, Udine, Italy. nataschab@libero.it.
9
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. t_barbara2001@yahoo.it.
10
Azienda Ospedaliero-Universitaria of Udine, P.le S. Maria della Misericordia 15, 33100, Udine, Italy. carloalberto.beltrami@uniud.it.
11
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. antonio.beltrami@uniud.it.
12
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. daniela.cesselli@uniud.it.
13
Clinic of Plastic and Reconstructive Surgery of Udine, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. piercamillo.parodi@uniud.it.
14
Azienda Ospedaliero-Universitaria of Udine, P.le S. Maria della Misericordia 15, 33100, Udine, Italy. piercamillo.parodi@uniud.it.

Abstract

INTRODUCTION:

Autologous fat grafting is commonly used to correct soft-tissue contour deformities. However, results are impaired by a variable and unpredictable resorption rate. Autologous adipose-derived stromal cells in combination with lipoinjection (cell-assisted lipotransfer) seem to favor a long-term persistence of fat grafts, thus fostering the development of devices to be used in the operating room at the point of care, to isolate the stromal vascular fraction (SVF) and produce SVF-enhanced fat grafts with safe and standardized protocols. Focusing on patients undergoing breast reconstruction by lipostructure, we analyzed a standard technique, a modification of the Coleman’s procedure, and three different commercially available devices (Lipokit, Cytori, Fastem), in terms of 1) ability to enrich fat grafts in stem cellsand 2) clinical outcome at 6 and 12 months.

METHODS:

To evaluate the ability to enrich stem cells, we compared, for each patient (n=20), the standard lipoaspirate with the respective stem cell-enriched one, analyzing yield, immunophenotype and colony-forming capacity of the SVF cells as well as immunophenotype, clonogenicity and multipotency of the obtained adipose stem cells (ASCs). Regarding the clinical outcome, we compared, by ultrasonography imaging, changes at 6 and 12 months in the subcutaneous thickness of patients treated with stem-cell enriched (n=14) and standard lipoaspirates (n=16).

RESULTS:

Both methods relying on the enzymatic isolation of primitive cells led to significant increase in the frequency, in the fat grafts, of SVF cells as well as of clonogenic and multipotent ASCs, while the enrichment was less prominent for the device based on the mechanical isolation of the SVF. From a clinical point of view, patients treated with SVF-enhanced fat grafts demonstrated, at six months, a significant superior gain of thickness of both the central and superior-medial quadrants with respect to patients treated with standard lipotransfer. In the median-median quadrant the effect was still persistent at 12 months, confirming an advantage of lipotransfer technique in enriching improving long-term fat grafts.

CONCLUSIONS:

This comparative study, based on reproducible biological and clinical parameters and endpoints, showed an advantage of lipotransfer technique in enriching fat grafts in stem cells and in favoring, clinically, long-term fat grafts.

Notes to consider:
A. Google search for:
“human adipose derived Stem Cells for lacrimal gland”
Will write another post specifically for this category this week.


B.
Comparison of Methods for Obtaining and Preparing Adipose Derived Stem Cells from Liposuction.

1. . 2011 May; 178(5): 2299–2310.

PMCID: PMC3081158

Adipose Tissue-Derived Stem Cell in Vitro Differentiation in a Three-Dimensional Dental Bud Structure

Francesco Curcio: ti.duinu@oicruc
Address reprint requests to Francesco Curcio, M.D., Department of Pathology and Experimental and Clinical Medicine, University of Udine, P.le S. Maria della Misericordia, 33100 Udine, Italy ti.duinu@oicruc

Materials and Methods

To isolate adipose tissue–derived stem cells (ASCs), nine raw human abdominal lipoaspirates, obtained with the informed consent of the donors (28 to 35 years old), were washed in PBS solution and then dissociated in Dulbecco’s modified Eagle’s medium (DMEM; Sigma-Aldrich, St. Louis, MO) containing 400 U/mL of collagenase type 2 (Wortington, Lakewood, NJ) for 15 minutes at 37°C. Samples were centrifuged for 15 minutes at 1,800 × g, then pellets were resuspended and filtered through a 70-μm pore-sized membrane. Filtered cells were plated into 100-mm dishes (2 × 106 cells per dish) in proliferation medium, derived from Gronthos et al, composed of F-12 Coon’s and Ambesi’s modified medium (Gibco-Invitrogen, Carlsbad, CA), Medium-199, and CMRL-1066 media (Sigma) supplemented with 1.25% type 0 human serum, 25 ng/mL of platelet-derived growth factor-bb, 25 ng/mL of epidermal growth factor, 25 ng/mL of insulin-like growth factor 1, 25 ng/mL of fibroblast growth factor-b (FGF-b; all from Immunotools, Friesoythe, Germany), 10−9 mol/L dexamethasone (MP Biomedicals, Solon, OH), 90 μg/L of linoleic acid (Sigma), 25 mg/L of ascorbic acid (Sigma), and 25 μg/mL of gentamicin (Gibco). Colonies developed in primary culture and reached near confluency in approximately 1 week. ASCs were maintained semiconfluent to prevent cell differentiation, and approximately 80% of the medium was replaced every 3 days. Primary human bone marrow mesenchymal stem cells (MSCs) and dental pulp stem cells (DPSCs) were obtained as described by Ferro et al. Human bone MSCs were obtained by flushing the femur head content through a 26-gauge needle. Flushed cells were diluted in Hanks’ balanced salt solution (Sigma), layered on top of Ficoll (Amersham), and centrifuged. Finally, buffy coat was washed twice with Hanks’ balanced salt solution and subjected to immunodepletion using RosetteSep (Stemcells Technologies, Vancouver, British Columbia). Then 1.5 × 106 of freshly isolated cells, derived from primary culture, were plated in 100-mm dishes (BD Falcon, San Jose, CA). Dental pulps were extracted from human deciduous teeth of 5- to 7-year-old children (with parents’ permission) using a syringe needle and were transferred to 35-mm Petri dishes (BD Falcon); human DPSC colonies developed in primary culture and usually reached confluence approximately 2 weeks later; both MSCs and DPSCs were made to proliferate in the same medium used for ASCs.
Human supranumeral teeth buds were isolated following the same methods used for DPSCs; were cultured in DMEM/F-12 (Gibco) containing 20% fetal bovine serum supplemented with 0.18 mg/mL of ascorbic acid (Sigma), 2 mmol/L L-glutamine (Sigma), and 50 U/mL of penicillin/streptomycin (Gibco); and were used as amelo-odontoblastic positive control cells. Human primary thyroid cells, used as negative controls in RT-PCR and immunoblot, were cultured according to the method of Curcio et al. Human embryonic carcinoma stem cells (Ntera2), used as a positive control for embryonic stem cell markers as suggested by Liedtke et al, were cultured in DMEM (Gibco), supplemented with 4.5 g/L of glucose, 4 mmol/L L-glutamine, 10% fetal bovine serum, and 50 U/mL of penicillin/streptomycin (Gibco).

2.


. 2011 May; 178(5): 2299–2310.
PMCID: PMC3081158

Adipose Tissue-Derived Stem Cell in Vitro Differentiation in a Three-Dimensional Dental Bud Structure

Federico Ferro, Renza Spelat, Giuseppe Falini, Annarita Gallelli, Federica D’Aurizio, Elisa Puppato, Maura Pandolfi, Antonio Paolo Beltrami, Daniela Cesselli, Carlo Alberto Beltrami, Francesco Saverio Ambesi-Impiombato,and Francesco Curcio


Note:
ADSC were obtained from human adipose tissue aspirates following a protocol optimized by Beltrami’s group for the isolation and in vitro expansion of human multipotent adult stem cells.20

As previously shown for multipotent adults stem cells obtained from human liver, bone marrow, heart and peripheral blood, ADSC expressed the pluripotent state-specific transcription factors Oct-4, Nanog and Sox 2 (Figure 1A–D) and were characterized by a mesenchymal stem cell immunophenotype. When assessed by flow-cytometry, ADSC highly expressed CD90, CD105, CD73, however, were mainly negative for the hematopoietic markers CD34 and CD45 (Figure 1E). Importantly, ADSC displayed multipotency, being able to differentiate into mature cell types of all the three germ layers. Specifically, when exposed to the proper differentiation inducing conditions, ADSC were able to give rise to endodermic (Figures 1F and G), mesodermic (Figures 1H and I) and ectodermic derivatives (Figures 1J and K).




3.   2013 Apr;38(4):451-63. doi: 10.3109/02713683.2012.763100. Epub 2013 Feb 1.

Human adipose-derived stem cells for the treatment of chemically burned rat cornea: preliminary results.

Author information

1
Department of Ophthalmology, Azienda Ospedaliero Universitaria Santa Mariadella Misericordia, University of Udine, Udine, Italy. markzeppieri@hotmail.com

Abstract

PURPOSE:

Adipose-derived stem cells (ADSC) are multipotent, safe, non-immunogenic and can differentiate into functional keratocytes in situ. The topical use of ADSC derived from human processed lipoaspirate was investigated for treating injured rat cornea.

METHODS:

A total of 19 rats were used. Six animals initially underwent corneal lesion experiments with 0.5 N NaOH (right eye) and 0.2 N (left). The 0.2 NaOH protocol was then used in 13 rats. All 26 eyes of 13 rats eyes received topical azythromycin bid for 3 d and divided into five treatment groups (n = 5 eyes/group), which included: control, stem cells, serum, stem + serum and adipose (raw human lipoaspirate). The four treatment groups received topical treatment three times daily for 3 d. Stem cells were isolated and harvested from human lipoaspirate. Topical eye drops were prepared daily with 1 × 10(5) cells/treatment. Fluorescein positive defect area and light microscope assessment was performed at 20, 28, 45, 50 and 74 h. Animals were sacrificed at 74 h for histological evaluation. Data were statistically analyzed for differences amongst groups.

RESULTS:

The stem cell-treated eyes had significantly smaller epithelial defects at each time point compared to control- and adipose-treated eyes (p < 0.05). This group showed slightly better epithelium healing than the serum and combined group, yet not significantly different. Histology showed that stem cell-treated corneas had complete re-epithelization, with less inflammatory cells and limited fibroblast activation structure compared with the control eyes.

Isolation and Preparation of Adipose-derived Stem Cells

Human subcutaneous abdominal adipose tissue was obtained from healthy patients (aged 27–62 years) undergoing elective lipoaspiration surgery with informed oral and written consent under a protocol approved by the Institutional Review Board (IRB) of the University of Udine, in accordance with the guidelines of the Tenets of the Declaration of Helsinki. Patients were screened and resulted negative for HIV, hepatitis B and C virus, and syphilis.
ADSC were obtained from the stromal vascular fraction (SVF) obtained from lipoaspirates and cultured as previously described.

Briefly, the SVF was obtained by centrifuging the lipoaspirates at 3000 g for 3 min. The SVF was subsequently dissociated in Jocklik modified Eagle’s medium (JMEM; Sigma-Aldrich, St Louis, MO) containing 400 U/mL of collagenase type 2 (Sigma-Aldrich) for 20 min at 37 °C. The collagenase enzymatic activity was stopped with the addition of 0.1% bovine serum albumin (BSA; Sigma-Aldrich) in JMEM. Samples were centrifuged for 10 min at 600g, then pellets were resuspended and filtered through a 40 µm pore-sized membrane. Filtered cells (2 × 106 cells per dish) were plated into 100 mm human fibronectin-coated (Sigma-Aldrich) dishes in an expansion medium composed as follows: 60% low glucose Dulbecco’s Modified Eagle Medium (DMEM; Invitrogen, Carlsbad, CA), 40% MCDB-201(Sigma-Aldrich), 1 mg/mL linoleic acid-BSA, 10−9 mol/L dexamethasone (MP Biomedicals, Solon, OH), 10−4 M ascorbic acid-2 phosphate (Sigma-Aldrich), 1Xinsulin-transferrin-sodium selenite (Sigma-Aldrich), 2% fetal bovine serum (FBS; StemCell Technologies, Vancouver, Canada), 10 ng/mL of human platelet-derived growth factor-bb and10 ng/mL of human epidermal growth factor (both from Peprotech EC, London, UK). Colonies developed in primary culture and reached near confluency in approximately 1 week. Medium was replaced every 3–4 d. Cells were detached with 0.25% trypsin–EDTA (Sigma-Aldrich) and replated at a density of 2 × 103/cm once reached at 70%–80% confluence. Adherent cells obtained after the second subculture, which corresponds to the third passage of cells, were used for the experiment.

Cells were isolated by the use of the selective medium. Stemness of these cells was demonstrated in vitro in accordance to our previous study on the basis of: mesenchymal stem cell-like surface immunophenotype; expression of Oct-4, Nanog and Sox2 proteins and multipotency, shown by the ability to differentiate into derivatives of all three germ layers.

Flow Cytometry

Proliferating cells were detached with 0.25% trypsin–EDTA (Sigma-Aldrich) and, after a 20 min recovery phase, were incubated with either properly conjugated primary antibodies: CD10, CD13, CD29, CD49a, CD49b, CD49d, CD90, CD73, CD44, CD59, CD45, CD271, CD34, (BD Biosciences, Le Pont-de-Claix, France), CD105, KDR, CD66e (Serotech, Kidlington, UK), CD133 (Miltenyi Biotec, Bergisch Gladbach, Germany), E-cadherin (Santa Cruz Biotechnology, Santa Cruz, CA), ABCG-2 (Millipore, Bioscience Research Reagents (formerly Chemicon), Temecula, CA). Properly conjugated isotype-matched antibodies were used as a negative control. The analysis was performed by CyAn (Dako Glostrup, Denmark).

Immunofluorescence

Cells cultured either in expansion or in differentiation medium were fixed in 4% buffered paraformaldehyde for 20 min at room temperature (R.T.). For intracellular stainings, fixed cells were permeabilized for 8 min at R.T. with 0.1% Triton X-100 (Sigma-Aldrich) before exposing them to primary antibodies. In order to block unspecific binding of the primary antibodies, cells were incubated with 10% donkey serum in PBS for 30 min. Primary antibody incubation was performed over-night at 4 °C using following dilutions: Oct-4 (Abcam, 1:150); Sox-2 (Millipore, Bioscience Research Reagents (formerly Chemicon), 1:150); Nanog (Abcam, 1:150); Cytokeratins 7, 8, 18, 19 (Biogenex, Freemont, CA, 1:20); ß3-tubulin (Abcam, 1:1000); Smooth Muscle Actin (Dako, 1:50), Connexin 43 (Santa Cruz, 1:40); α-Sarcomeric Actin (Sigma, 1:100) and Gata4 (Santa Cruz, 1:100). To detect primary antibodies, A488 and A555 dyes labeled secondary antibodies, diluted 1:800, were employed (Molecular Probe, Invitrogen). 0.1 μg/mL DAPI (Sigma-Aldrich) was used to identify nuclei. Vectashield (Vector, Burlingame, CA) was used as a mounting medium. Epifluorescence images were obtained utilizing a live cell imaging dedicated system consisting of a Leica DMI 6000B microscope connected to a Leica DFC350FX camera (Leica Microsystems, Wetzlar, Germany).

Multilineage Differentiation

Hepatocytic differentiation was induced for growing cells for 2 weeks at high density onto fibronectin-coated dishes in a medium containing 0.5% FBS, 10 ng/mL FGF-4 and 20 ng/mL HGF (both from Peprotech EC). After this period, FGF-4 and HGF were substituted for 20 ng/mL OncostatinM for another 14 d (Peprotech EC). Muscle cell differentiation was achieved by plating 0.5 to 1  ×  104/cm2 cells in an expansion medium containing 5% FCS (Sigma-Aldrich), 10 ng/mL bFGF, 10 ng/mL VEGF and 10 ng/mL IGF-1 (all from Peprotech EC), but not EGF. Cells were allowed to become confluent and cultured for up to 4 weeks with medium exchanges for every 4 d. For neurogenic differentiation, cells were plated in DMEM-high glucose (Invitrogen), 10% FBS (Sigma-Aldrich). After 24 h medium was replaced with DMEM-high glucose, 10% FBS containing B27 (Invitrogen), 10 ng/mL EGF and 20 ng/mL bFGF (both from Peprotech EC). After 5 d, cells were washed and incubated with DMEM containing 5 g/mL insulin, 200 µM indomethacin and 0.5 mM IBMX (all from Sigma-Aldrich) in the absence of FBS for 5–10 d. At the end of every treatment, cells were fixed with 4% buffered paraformaldehyde.

Experiment for Detecting the Presence of Stem Cells

The preparation of ADSC for treatment was performed in a similar manner to the previous experiments. The cells were allowed to grow until 80% confluence and detached with the trypsin solution, centrifuged and pelleted. The supernatant was discarded and resuspended in a labeling solution containing 25 µm of CFSE in sterile PBS (0.1 M pH 7.4). The cells remained in incubation in this medium for 15 min, then incubated for an additional 30 min with a fresh medium. Cells were washed and centrifuged, then resuspended in HBSS to a final concentration of 5.0 × 106 cells/mL. A small fraction of the cells were plated in a culture dish to demonstrate the viability of the cells after the labeling and the effectiveness of the fluorescent staining.
After 12 h of treatment and one night of rest (24 h after injury), the rat was scarified by an overdose of sodium pentobarbital. Both eyes were enucleated and fixed by immersion in Somogyi’s fixative (4% (w/v) paraformaldehyde and 15% (v/v) saturated picric acid solution in phosphate buffer 0.1 M).
The eyeglobes were then cryopreserved in 30% sucrose solution for 2 h and rapidly frozen in liquid nitrogen. We then obtained 5 µm sections on a cryostat microtome. The sections were counterstained with DAPI to label the nuclei and mounted with fluorescent mounting medium (DAKO).

Other Notes:

1.

 2015 Jan 5;6:2. doi: 10.1186/scrt536.

Adipose tissue derived stem cells: in vitro and in vivo analysis of a standard and three commercially available cell-assisted lipotransfer techniques.

Author information

1
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. rossana.domenis@uniud.it.
2
Clinic of Plastic and Reconstructive Surgery of Udine, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. laralazzaro@hotmail.com.
3
Clinic of Plastic and Reconstructive Surgery of Udine, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. sarah2012@hotmail.com.
4
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. damianomng@gmail.com.
5
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. byoteck@libero.it.
6
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. jenny_bourkoula@hotmail.com.
7
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. ivana.manini@tiscali.it.
8
Azienda Ospedaliero-Universitaria of Udine, P.le S. Maria della Misericordia 15, 33100, Udine, Italy. nataschab@libero.it.
9
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. t_barbara2001@yahoo.it.
10
Azienda Ospedaliero-Universitaria of Udine, P.le S. Maria della Misericordia 15, 33100, Udine, Italy. carloalberto.beltrami@uniud.it.
11
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. antonio.beltrami@uniud.it.
12
Department of Medical and Biological Sciences, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. daniela.cesselli@uniud.it.
13
Clinic of Plastic and Reconstructive Surgery of Udine, University of Udine, P.le Kolbe 4, 33100, Udine, Italy. piercamillo.parodi@uniud.it.
14
Azienda Ospedaliero-Universitaria of Udine, P.le S. Maria della Misericordia 15, 33100, Udine, Italy. piercamillo.parodi@uniud.it.

Abstract

INTRODUCTION:

Autologous fat grafting is commonly used to correct soft-tissue contour deformities. However, results are impaired by a variable and unpredictable resorption rate. Autologous adipose-derived stromal cells in combination with lipoinjection (cell-assisted lipotransfer) seem to favor a long-term persistence of fat grafts, thus fostering the development of devices to be used in the operating room at the point of care, to isolate the stromal vascular fraction (SVF) and produce SVF-enhanced fat grafts with safe and standardized protocols. Focusing on patients undergoing breast reconstruction by lipostructure, we analyzed a standard technique, a modification of the Coleman’s procedure, and three different commercially available devices (Lipokit, Cytori, Fastem), in terms of 1) ability to enrich fat grafts in stem cellsand 2) clinical outcome at 6 and 12 months.

METHODS:

To evaluate the ability to enrich stem cells, we compared, for each patient (n=20), the standard lipoaspirate with the respective stem cell-enriched one, analyzing yield, immunophenotype and colony-forming capacity of the SVF cells as well as immunophenotype, clonogenicity and multipotency of the obtained adipose stem cells (ASCs). Regarding the clinical outcome, we compared, by ultrasonography imaging, changes at 6 and 12 months in the subcutaneous thickness of patients treated with stem-cell enriched (n=14) and standard lipoaspirates (n=16).

RESULTS:

Both methods relying on the enzymatic isolation of primitive cells led to significant increase in the frequency, in the fat grafts, of SVF cells as well as of clonogenic and multipotent ASCs, while the enrichment was less prominent for the device based on the mechanical isolation of the SVF. From a clinical point of view, patients treated with SVF-enhanced fat grafts demonstrated, at six months, a significant superior gain of thickness of both the central and superior-medial quadrants with respect to patients treated with standard lipotransfer. In the median-median quadrant the effect was still persistent at 12 months, confirming an advantage of lipotransfer technique in enriching improving long-term fat grafts.

CONCLUSIONS:

This comparative study, based on reproducible biological and clinical parameters and endpoints, showed an advantage of lipotransfer technique in enriching fat grafts in stem cells and in favoring, clinically, long-term fat grafts.


More Notes: 
 4.5. Angiogenesis Regulation
Angiogenesis is defined as the process by which new vasculature sprouts from pre-existing blood
vessels. Normal angiogenesis is important during wound healing process. Various studies have
demonstrated the effect of MSC secretome on key steps in angiogenesis. For example, different MSC
populations (e.g., adipose, amniotic, bone marrow (BM) and Wharton jelly umbilical vein) induce
proliferation and migration of endothelial cells promoting tube formation, as well as prevent
endothelial cell apoptosis in vitro [109].

From:
International Journal o f
Molecular Sciences
Review
Mesenchymal Stem Cell Secretome: Toward Cell-Free
Therapeutic Strategies in Regenerative Medicine
Francisco J. Vizoso 1,*, Noemi Eiro 1, Sandra Cid 1, Jose Schneider 2 and
Roman Perez-Fernandez 3,*
1 Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33290 Gijón, Spain;
noemi.eiro@gmail.com (N.E.); investigacion@hospitaldejove.com (S.C.)
2 Department of Obstetrics & Gynecology, C/Ramon y Cajal 7, University of Valladolid, 47005 Valladolid,
Spain; jose.schneider@urjc.es
3 Department of Physiology-Center for Research in Molecular Medicine and Chronic Diseases (CIMUS),
University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
* Correspondence: franvizoso@gmail.com (F.J.V.); roman.perez.fernandez@usc.es (R.P.-F.);
Tel.: +34-985-320-050 (F.J.V.); +34-881-815-421 (R.P.-F.); Fax: +34-985-315-710 (F.J.V.)
Received: 28 July 2017; Accepted: 22 August 2017; Published: 25 August 2017
Abstract: Earlier research primarily attributed the effects of mesenchymal stem cell (MSC) therapies to
their capacity for local engrafting and differentiating intomultiple tissue types.However, recent studies
have revealed that implanted cells do not survive for long, and that the benefits of MSC therapy
could be due to the vast array of bioactive factors they produce, which play an important role in the
regulation of key biologic processes. Secretome derivatives, such as conditioned media or exosomes,
may present considerable advantages over cells for manufacturing, storage, handling, product shelf
life and their potential as a ready-to-go biologic product. Nevertheless, regulatory requirements for
manufacturing and quality control will be necessary to establish the safety and efficacy profile of
these products. Among MSCs, human uterine cervical stem cells (hUCESCs) may be a good candidate
for obtaining secretome-derived products. hUCESCs are obtained by Pap cervical smear, which is
a less invasive and painful method than those used for obtaining other MSCs (for example, from bone
marrow or adipose tissue). Moreover, due to easy isolation and a high proliferative rate, it is possible
to obtain large amounts of hUCESCs or secretome-derived products for research and clinical use.
Keywords: conditioned media; exosomes; mesenchymal stem cells; adipose-derived stem cells;
bone marrow mesenchymal stem cells; uterine cervical stem cells; hUCESCs

Sandra Lora Cremers, MD, FACS
Johns Hopkins University Medicine, Suburban Hospital

Office: 301-896-0890
*One Central Plaza. 11300 Rockville Pike, Suite 1202. Rockville, MD 20852
*Van Ness Center. 4301 Connecticut Ave., NW, Suite 125. Washington, DC 20008
www.voeyedr.com
eyedoc2020.blogspot.com


Attention:  This communication may contain protected health information (PHI) that is legally protected from inappropriate disclosure by the Privacy Standards of the Health Insurance Portability Act (HIPAA) and relevant Maryland Laws.  If you are not the intended recipient, please note that any dissemination, distribution or copying of this communication is strictly prohibited.  If you have received this message in error, you should notify the sender immediately by telephone or by return e-mail and delete this message from your computer.

Shopping Cart