Review Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Aug 26, 2017; 9(8): 107-117
Published online Aug 26, 2017. doi: 10.4252/wjsc.v9.i8.107
Adipose-derived stromal cell in regenerative medicine: A review
Reza Tabatabaei Qomi, Mohsen Sheykhhasan, Department of Stem Cell, the Academic Center for Education, Culture and Research, PO Box QOM-3713189934, Qom, Iran
Author contributions: Tabatabaei Qomi R performed the majority of the writing prepared the figures and tables; Sheykhhasan M performed data accusation and writing; Tabatabaei Qomi R provided the input in writing the paper; Sheykhhasan M designed the outline and coordinated the writing of the paper.
Conflict-of-interest statement: There is no conflict of interest associated with any of the senior author or other coauthors contributed their efforts in this manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Mohsen Sheykhhasan, MS, Department of Stem Cell, the Academic Center for Education, Culture and Research, Qom Branch, PO Box QOM-3713189934, Qom, Iran. m.sheykhhasan@acecr.ac.ir
Telephone: +98-253-2700152 Fax: +98-253-2700155
Received: January 27, 2017
Peer-review started: February 12, 2017
First decision: March 10, 2017
Revised: April 30, 2017
Accepted: May 18, 2017
Article in press: May 19, 2017
Published online: August 26, 2017

Abstract

The application of appropriate cell origin for utilizing in regenerative medicine is the major issue. Various kinds of stem cells have been used for the tissue engineering and regenerative medicine. Such as, several stromal cells have been employed as treat option for regenerative medicine. For example, human bone marrow-derived stromal cells and adipose-derived stromal cells (ADSCs) are used in cell-based therapy. Data relating to the stem cell therapy and processes associated with ADSC has developed remarkably in the past 10 years. As medical options, both the stromal vascular and ADSC suggests good opportunity as marvelous cell-based therapeutics. The some biological features are the main factors that impact the regenerative activity of ADSCs, including the modulation of the cellular immune system properties and secretion of bioactive proteins such as cytokines, chemokines and growth factors, as well as their intrinsic anti-ulcer and anti-inflammatory potential. A variety of diseases have been treated by ADSCs, and it is not surprising that there has been great interest in the possibility that ADSCs might be used as therapeutic strategy to improve a wider range of diseases. This is especially important when it is remembered that routine therapeutic methods are not completely effective in treat of diseases. Here, it was discuss about applications of ADSC to colitis, liver failure, diabetes mellitus, multiple sclerosis, orthopaedic disorders, hair loss, fertility problems, and salivary gland damage.

Key Words: Adipose-derived stromal cell, Colitis disease, Liver failure, Diabetes mellitus, Multiple sclerosis, Orthopedic disorders, Hair loss, Fertility problems, Salivary gland damage

Core tip: Nowadays, adipose-derived stromal cells (ADSCs) are one of the most important and promising cell sources in the field of regenerative medicine. Unique capabilities of ADSCs caused them to be used in both research and treatment as a valuable resource in basic science and medical researches. In over 15 years since their discovery, ADSCs have transformed our toolkit for treating human disorder and disease. As the field enters its next decade, a new wave of therapeutic applications, such as hepatic regeneration, diabetes mellitus treatment, multiple sclerosis treatment, and orthopaedic disorders regeneration, has converged with ADSCs to yield new insights for their use in stem cell engineering and regenerative medicine.



INTRODUCTION

Mesenchymal stromal cells (MSCs) are undifferentiated cells that are able to renew their population and become differentiated to produce all specialized cell types of the tissue from which they are originated[1] (Figure 1). While MSCs are traditionally isolated from bone marrow, over the last few years, they have also been found in many other adult tissues such as liver, cord blood, placenta, dental pulp and adipose tissue[1]. The different stromal cells have some features in common, including morphological and immunophenotypic properties[1]. Although, bone marrow-derived stromal cells (BMSCs) and adipose-derived stromal cells (ADSCs) are better known than others[2-4]. ADSCs share biological properties with stromal cells obtained from bone marrow; however, these candidate cells also have some different properties compared to BMSCs[2,3].

Figure 1
Figure 1 Schematic demonstration of the Biological Properties of human adipose-derived stromal cells. ADSC: Adipose-derived stromal cell.

Furthermore, Both Adipose-derived stromal cells and bone marrow-derived stromal cells have played a prominent role in regenerating the defective tissue of patients[4]. In the recent years, as one of the most successfully developed stem cells, adipose-derived stromal cell is a better choice than many other adult stem cells such as bone marrow-derived stromal cell because of its characters[4].

Such as, ADSCs not only have decreased sampling risk for individual donors compared with BMSCs but also have been needed to an easier method for isolation compared with BMSCs[4]. The adipose tissue, ADSCs’ harvested source, could also provide a higher number of stromal cells compared with bone marrow tissue as BMSCs’ obtained source[4]. Furthermore, ADSCs are superior to BMSCs in some biological features, including the immune feature regulation[4]. In addition, with an emphasis on adult stem cells rather than on embryonic stem cells, regenerative medicine programs are using ADSCs as more applicable adult stem cells to treat different diseases[4]. Today, Adipose-derived stromal cells are known as a rich source of MSCs which are considered a suitable case for repair and regeneration of various tissues because of their rapid proliferation and multilineage potential[2-5]. Several properties making scientists to pay attention to ADSCs include Immunomodulatory effects and secretion of a variety of growth factors and cytokine as well as anti-apoptosis and anti-inflammation potential[6]. In vitro ADSCs are identified by plastic adherence, colony forming capacity, rapid proliferation and lack of major histocompatibility class II (MHCII)[7,8]. ADSCs not only are interesting in basic sciences, but also have been used in a broad range of regenerative medicine application, such as orthopaedic damage, fertility problems, hair loss, Colitis disease, liver failure, diabetes mellitus, multiple sclerosis, etc. In the treatment of many of different diseases, ADSCs have exhibited a great potential for tissue repair and modulation of host immune response in vivo[6]. ADSCs from healthy donors are an attractive cell source for organ regeneration[9]. These cells can be obtained and cultured in vitro in sufficient numbers and subsequently used in damaged tissue regeneration[10]. So far it has been well recognized that these cells possess a broad spectrum of differentiated potentials, from cell types of mesodermal origin to ectoderm (such as hepatocyte) and endoderm (such as beta cells), when induced in vitro[10-13] (Figure 2). ADSC can be expanded effortlessly in culture for long periods of time without losing their differentiation capacity[12]. They are robust cells, which can easily survive freezing temperatures with limited loss in viability, proliferative capacity and differentiate potency[12]. The most attractive aspect of ADSCs is their immunosuppressive properties that allow transplanting them irrespective of a human leukocyte antigen (HLA) match between the host and the donor[14]. These cells are negative for surface marker proteins, such as CD14, CD34, and CD45, in vitro[15]. Although, these candidate cells express CD34 in vivo[16].

Figure 2
Figure 2 Schematic demonstration of the multidifferentiation potential of human adipose-derived stromal cells in vitro. ADSCs: Adipose-derived stromal cells.

Moreover, ADSCs express cell surface markers, including CD10, CD13, CD29, CD44, CD71, CD73, CD90, CD105, CD166 and CD271 (Figure 3) and different varieties of trophic factors, such as molecular regulation of cell growth and proliferation, fibrosis, angiogenesis, and immune suppression[7,17-24]. Additionally, the Anti-apoptotic, anti-oxidant, anti-inflammatory activities of the ADSCs are among other important characteristics that can affect theirs regenerative potential[9,25-29].

Figure 3
Figure 3 Schematic illustrations of the surface markers of freshly isolated human adipose-derived stromal cells. ADSCs: Adipose-derived stromal cells.

Furthermore, ADSC treatment is now a widely used therapeutic strategy in the field of medicine because of its intrinsic therapeutic properties, relatively easy approach to harvesting them, and the large number these cells obtained after isolation[29].

In 160 clinical trials, ADSC-based therapy has been also used to treat various diseases such as orthopaedic disorders, hepatic failure, inflammatory diseases, and autoimmune disease[30].

SAFETY ISSUES OF ADSC CELLS

The safety study of ADSCs conducted by the different preclinical and clinical trial has documented that these candidate cells are safe enough to be used in various treatment methods and can also play an effective role in the treatment of diseases[31-42]. Such as, the current finding has shown that autologous ADSCs could act as a safety agent in muscle defect regeneration, both smooth and skeletal muscle, due to their profibrotic properties as well as trophic factors[43]. Based on the results obtained from a clinical trial study, it was verified that used adipose-derived stromal cell (ADSC) implantation showed an appropriate safety feature with no serious complication in patients with degenerative disc disease[44]. Furthermore, in phase II of the clinical trial study, the ADSC injections into the knee of 18 patients with osteoarthritis (OA) showed that these procedures do not have any severe adverse effects[45].

However, there is little report about severe adverse effects. Such as, some of the adverse effects observed during the study include headache, inflammation, etc.[46]. Furthermore, the safe use of ADSCs in cosmetic reconstructive surgery following a tumor is particularly doubtful because of the potential of these candidate cells to promote the development and progression of cancer[47,48].

These cell candidates could be further assessed for understanding their therapeutic potential and safety issues in them.

In review study, we focused on ADSCs application in treat of inflammatory disease, liver failure, complication related to diabetes mellitus, multiple sclerosis diseases, orthopaedic disorders, hair loss, fertility problems, and salivary gland damage, both in vivo and clinical study. Also, it was provided the significant number of ADSC-based clinical trials (Table 1)[35-41,45,49-55].

Table 1 List of clinical trials that use stromal vascular fraction or adipose-derived stromal cells for a variety diseases treatment.
Type of cellsClinical trial phaseDiseaseNo. of patientHighlight findingRef.
Autologous ADSCsPhase IAmyotrophic lateral sclerosis27 patientsLow back and radicular leg pain were observed, no tumor formation were observed[35]
Stromal vascular fractionPhase 0 (CSN111)Peyronie’s disease11 patientsNo serious adverse events were observed[36]
Autologous adipose-derived stromal vascular fractionPhase IImpaired hand function in patients with systemic sclerosis12 patientsFour minor adverse events were observed, hand disability and pain were decreased[54]
Autologous adipose-derived stromal vascular fractionPhase I (NTC01813279)Impaired hand function in patients with systemic sclerosis12 patientsMobility, strength and fibrosis of the hand was improved[55]
Autologous adipose-derived stromal vascular fractionPhase 0Systemic sclerosis12 patientsFinger oedema, skin sclerosis, motion and strength of the hands were ameliorated, hand pain was decreased[49]
Stromal vascular fraction combined with PRPPhase 0 (NCT02097862)Degenerative disc disease15 patientsNo serious adverse events were observed[37]
Autologous ADSCsPhase I/IIOsteoarthritis18 patientsAdverse events were observed in several patients, including urinary stone, arthralgia, pain and tenderness in the pes anserinus of the ipsilateral knee; ADSCs injection into the osteoarthritic knee ameliorated function and pain of the knee joint[45]
ADSCPhase 0 (NCT02357485)Osteoarthritis6 patientsNo serious adverse events were observed, pain for osteoarthritis of the knee was decreased[38]
Stromal vascular fraction containing ADSCsPhase 0Osteochondral lesions of the talus49 patientsSVF Containing ADSCs administration was a therapeutically beneficial strategy for osteochondral lesions of the talus[50]
Expanded ADSCsPhase 0Patients with a desire to become pregnant (with Crohn's Perianal Fistula)6 patientsLocal administration of ADSCs did not impact on course of pregnancy or newborn development;[39]
Adipose-derived mesenchymal stem cellPhase I/II (NCT02513238)Salivary gland hypofunction and radiation-induced xerostomia30 patientsNo serious adverse events were observed Change in unstimulated whole salivary flow rate was observed; no serious adverse events were observed[40]
ADSCPhase 0Alopecia20 patientsHair diameter and density were improved,the efficacy and the safety of the treatment with ADSCs was confirmed[51]
Conditioned media of adipose tissue-derived stem cellsPhase 0Female pattern hair loss27 patientsHair density was enhanced, hair thickness was improved[52]
ADSC conditioned mediumPhase 0Alopecia22 patientsHair numbers were considerably enhanced[53]
Expanded autologous ADSCPhase 0Type 2 diabetes mellitus3 patientsBlood glucose levels were decreased in all patients, no serious adverse events were observed[41]
COLITIS DISEASE

Colitis, an inflammation of the colon, was treated with using intraperitoneal injection 105-106 human ADSCs or murine ADSCs in a study[56,57]. This study was associated with reduced weight loss, improved survival and improved clinical in ADSC groups[57]. In another study, intravenous tail vein administration of 106 macrophages cultured with either human ADSCs vs mouse ADSC lead to ameliorated disease activity index, alleviated weight loss and mortality in mice treated with ADSCs and ADSC-MF (macrophages cultured with ADSCs)[58]. Also, intraperitoneal infusion 2 × 106 human and mouse ADSCs demonstrated significant attenuate in inflammation scores overall the colon and increase weight[59].

LIVER FAILURE

The liver is a complicated organ that plays a metabolic function in human body. Any damage to this vital organ causes irreparable damage in the body. Due to this fact that adipose-derived stromal cells can differentiate into hepatocyte-like cells, both in vitro and in vivo condition, as well as capabilities such as homing in the defect location, and immunomodulatory and anti-apoptotic mechanism, they are used for liver failure treatment[29,60,61]. Furthermore, these cells are including anti-inflammatory factors and secrete various factors involved in tissue regeneration and are considered as a new therapeutic strategy to rebuild of liver damage[60,62].

Previous studies have display that ADSC transplantation demonstrates appropriate therapeutic outcomes for multiple diseases, including liver failure[63-65]. It is cleared that human ADSC transplantation could efficiently improve the liver function of acute liver failure (ALF) rats[66]. Furthermore, ADSCs administration increased the survival rates as well as decreased the ALF conditions in an immunocompetent ALF rat model[67].

DIABETES MELLITUS

Diabetes mellitus, a multifactor disease, is one of the main factors of death around the world. Because of the regenerative capacity and growth factors, cytokines, and chemokines secretion, in addition to angiogenesis and vascularization features, stromal vascular fraction has suitable potential for the therapeutical application in major complication of diabetes mellitus including foot ulcer related to diabetic, nephropathy and retinopathy[68]. An experiment on diabetes athymic rat illustrate that ADSCs injection to vascular network of retina dysfunction site can significantly decrease apoptosis and vascular leakage and increase vascular synthesis and attenuate neurodegeneration[69].

MULTIPLE SCLEROSIS

Multiple sclerosis, one of the most devastating autoimmune diseases of the nervous system, can be found throughout the entire world[70]. Several animal studies have been performed on this disease using ADSC and stromal vascular fraction (SVF)[68,71-75]. In other studies the beneficial effects of ADSC and SVF have been evaluated on experimental autoimmune encephalitis (EAE), another disease of the nervous system[76,77]. One such animal study indicated that SVF may also have a therapeutic effect on multiple sclerosis[76]. In another study, the use of both ADSC and SVF resulted in a reduction in the demyelination and pathological features of EAE[78]. Both of these studies demonstrate that SVF, when employed in combination with ADSC, can lead to an amelioration of EAE in a murine model[78]. In one study, the expression level of interleukin-10 as an immunomodulator factor was high[78]. Additionally, an in vivo study identified that an ADSC-conditioned medium, along with ADSC, has both neuroprotective and immunomodulatory effects, suggesting the use of this conditioned medium as a valuable agent for treatment of EAE[79]. Meanwhile, neither pre-clinical results nor clinical evidence have demonstrated any serious adverse effects of ADSC administration[75-78]. In one clinical study, four patients with multiple sclerosis were treated using ADSC injection[75]. The clinical outcome demonstrated that ADSC administration is an effective treatment strategy for patients with multiple sclerosis[75]. Moreover, the murine EAE model has demonstrated that ADSC may be used to ameliorate motor function and decrease inflammation[76].

Moreover, in a phase I dose-escalation safety trial noted that intrathecal treatment of autologous adipose-derived stromal cells appears safe at the tested doses in amyotrophic lateral sclerosis[35]. Compared to use of fat transplantation, use of ADSCs in systemic sclerosis (SS) patients improved mouth functional disability, demonstrating the importance of ADSCs administration in patients suffering from SS[80].

ORTHOPAEDIC DISORDERS

Orthopaedic disorders have been considered as leading problems in the human community.

Since ADSCs contain therapeutic properties (i.e., differentiation capability into a variety of cell lineage in vitro as well as having immunosuppressive, osteoinductive and anti-inflammatory features), they might be used for treatment of orthopedic major diseases such as degenerative OA[81,82].

It was reported that ADSCs increased the expression of osteogenic genes [i.e., runt related transcription factor 2 (RUNX2), Alkaline phosphatase, Type I collagen] and chondrogenic genes [i.e., Type II collagen, SRY-box 9 (SOX9) and aggrecan] on biomaterials in a chondrogenic inducing medium[83].

Previous studies showed that administration of both ADSCs and SVF in early OA is a safe and therapeutically efficient approach[82-86].

A study on rabbit model indicated that an eight week ADSCs/hydroxyapatite implantation to critical size tibial defects could remarkably enhance mineral content and bone regeneration[87]. Additionally, two clinical trials on bone healing illustrated that ADSCs in combination with synthetic bone graft and biomaterials may affect the regeneration, augmentation and vascularization of bone fracture[88].

Injection of ADSCs via second-look arthroscopy improved cartilage regeneration and decreased pain in patients with OA[89]. In addition, Jo et al[45] (2014) reported that the injection of 1 × 108 cell/mL ADSCs improved degenerative OA of 18 patients histologically and clinically after 6 mo of injection.

HAIR LOSS

Hair loss is one of the most crucial cosmetic challenges in both women and men these days. It’s a problem for the young and old alike.

ADSCs have great potential in hair repair and regeneration, so they are an important option for hair loss treatment[51,53,90-93]. The paracrine characteristics of ADSCs may include the specific factors released by them, including VEGF, HGF, IGF, and PDGF, which exert the specified effects on hair loss regeneration[51-53,93-95]. These factors are too therapeutically appropriated to be used for clinical application in patients with hair loss[51-53,93-95]. According to recent studies, it has been found that using ADSCs can stimulate hair growth in animal models[93]. Studies have shown that the conditioned medium (CM) derived from ADSCs also had proliferative effects on hair cells in vitro[91]. It was declared that a conditioned medium of ADSCs could lead to hair regeneration by promoting hypoxia[96]. Furthermore, a clinical trial involving 22 participants with alopecia documented that intradermal administration of a conditioned medium of ADSCs may lead to an ameliorating effect in hair regeneration process[53].

These medium candidates, in combination with LL-37, could also induce hair regeneration in vivo[90]. Furthermore, a study demonstrated that ADSCs-conditioned medium not only has a stimulated alkaline phosphatase activity, but is also related to dermal papillae cells and dermal papillae markers[97].

In addition, a retrospective observational study noted that hair density and thickness could be improved following 12 wk of CM-ADSC administration[52].

Previous evidence has demonstrated that ADSCs and adipocytes could act as a niche for hair follicles, due to providing an increase in the skin’s thickness and progress in the intradermal adipocyte layer during the anagen phase, as well as creating a decrease in the intradermal adipocyte layer during the catagen and telogen phases[98,99]. In addition, ADSCs and adipocytes regulate the hair cycle via the release of signaling molecules, i.e., WNTs, PDGF, BMPs, and FGFs[98,99]. These signals could lead to activation of the stem cell differentiation in the hair follicle and bulge stem cell activation during the telogen phase[98,99]. Canine ADSCs administration could also be caused by the increase in the vascularization process in the dermal papillae and has a beneficial effect on hair growth and repair in the nude mice model[100].

In addition, an animal study showed that ADSCs in combination with core-shell sphere could help in the formation of hair[97].

ADSCs can also support auditory hair cells, and these cells are capable of regenerating damaged hair[101].

Furthermore, protein secreted by ADSCs may be considered an appropriate tool for hair repair[93]. Such as, an observational pilot experiment performed on twenty seven patients with female pattern hair loss demonstrated that administration of protein extract derived from ADSCs could be caused to enhance in hair density and thickness since 12-wk follow up treatment. Furthermore, no serious complication was observed in patients[93].

Similarly, another pilot experiment verified the beneficial effect of ADSCs protein extract in patients with male pattern hair loss[93].

FERTILITY PROBLEMS

Infertility is one of the most common problems impacting both men and women. This health issue could lead to decreased populations, and treatment strategies are necessary to address it. However, many current therapeutic strategies are not very effective. As a result, more efficient treatments should be developed. One such solution is ADSCs-based therapy, which has been demonstrated to lead to improvements in fertility rates.

An animal study illustrated that the administration of ADSC could be considered as a therapeutic strategy in chemotherapy-induced ovarian dysfunction in rat models[102].

For example, the use of ADSCs caused a significant increase in the number of maturing follicles and corpora lutea with definite oocytes inside[103].

In another preclinical experiment, the use of collagen scaffold in combination with ADSCs enhances the short-term maintenance of ADSCs in ovaries[104].

It aims for long-term recovery of ovarian function, in addition to improving the fertility of rats with premature ovarian failure[104].

Similarly, Sun et al[105] (2013) demonstrated that intraperitoneal injection of ADSCs could ameliorate ovarian function in mice with chemotherapy-induced ovary damage.

Furthermore, on considering an in vitro study, it was identified that conditioned medium obtained from ADSCs could lead to human oocyte maturation and embryo formation following intracytoplasmic sperm injection through secretion of paracrine factors[106].

It is elucidated that the administration of ADSCs could promote fertility restoration in azoospermia rats, as well as the generation of sperm in them[107].

In addition, a human in vitro study reported that supernatant product of ADSCs (SPAS) could be ameliorate sperm motility in male infertile patients that can be due to existence of bioactive molecules and growth factors, which have a positive effect on sperm motility parameter[108].

In several animal studies, it was identified that ADSCs injection or transplantation have a positive impact on the viability of ovarian follicles and could increase the retention of short-term cryopreserved ovarian grafts, as well as improve the graft quality in the rat model[102-105,109].

Several preclinical and in vivo experiments have verified that the administration of ADSCs may have a beneficial effect on Peyronie’s disease, which is a problem that could lead to infertility[110-113].

Considering obtaining data from a pilot study, the application of autologous SVF in combination with shock wave may have a therapeutic effect on Peyronie’s disease[36].

In addition, this study on 11 patients documented that the administration of these adult stem cells is a safe process for the treatment of Peyronie’s disease[36].

SALIVARY GLAND DAMAGE

The salivary gland is considered as one of the most exocrine glands that generate saliva, which helps the chewing and swallowing process.

Radiotherapy is one of the most well-known agents that may cause damage to the salivary gland.

Due to their capacity to differentiate into salivary gland cells and their potential to secrete bioactive molecules, as well as their capability to induce regeneration following salivary gland failure, ADSCs have been considered promising tools for salivary gland damage regeneration[114-124].

There are a number of researches and clinical studies which have looked into salivary gland damage regeneration through ADSC application, by means of local and systemic use[114-124].

It was demonstrated that systemic use of ADSCs could provide a support against salivary gland damage induced by irradiation[114]. In addition, it was identified that ADSCs may migrate engrafting to an injured location via the blood stream[114,120,121].

Similarly, Maria et al[119] (2011) documented that ADSCs non-permanently supply a salivary gland cell of the endothelial or salivary acinar cell phenotypic trait by transdifferentiation into salivary gland cells.

It was identified that ADSC administration may be decreased in the apoptosis process through secreted growth factor with anti-apoptotic action[114,116,117,120,122]. Furthermore, the fibrosis reaction was diminished after ADSC administration[114,120]. It has been proposed that a paracrine mechanism could be responsible for the improvement of induced damage by radiation through providing growth factors related to neovascularization[120].

Both animal and human experiments have verified that ADSCs could represent a safe treatment strategy for salivary gland damage[40,114,115,117,120,123].

In addition, secreted bioactive factors from ADSCs could promote epithelial proliferation and a stimulated angiogenesis process[114,120,124].

One study explored the local administration of ADSCs for improved tissue remodeling effectiveness in impaired salivary glands induced by radiation[117]. This study showed that ADSCs could lead to beneficial results by ameliorating the tissue remodeling of impaired salivary glands induced by radiation[117].

Furthermore, it was elucidated that ADSC secretome from a hypoxic-conditioned medium may provide a positive outcome on radiation damaged salivary glands, as well as supplying ameliorating and remodeling effects on damaged tissue by paracrine mechanisms[118].

It was noted that ADSC application could also amend xerostomia induced by radiation, a problem related to salivary dysfunction that it is created following radiotherapy for head and neck cancer, through high expression of a variety of growth factors, including hepatocyte growth factor, and vascular endothelial growth factor[40,120,121].

CONCLUSION

Cell-based therapy has been used during the recent years to treat a variety of body damages and lesions. A variety of stromal stem cells harvested from several different tissue types have therapeutic characteristics, but BMSCs and ADSCs are widely considered more usable candidates for regenerative medicine among them. The application of ADSCs is greater than that of BMSCs in regenerative medicine because ADSCs have need to more easily technique for isolation compared to BMSCs, as well as they have a much greater rate in number than to BMSCs. because the technique for isolating ADSCs is easier and, consequently, they can be used in greater number than BMSCs.

As a result of their inherent therapeutic properties, ADSCs could also provide a hopeful strategy in the field of regenerative medicine for treatment a wide range of diseases and lesions. This will ensure the availability of ADSCs for research, trial and clinical applications in the future. Due to the promising results obtained from preclinical and clinical trials as well as their unique features in term of regenerative potential, these cells can be useful in the treatment of different diseases. Furthermore, it has been shown that the administration of ADSCs can provide a safe treatment strategy in regenerative medicine approaches. There have been few reported serious side effects resulting from the clinical use of ADSCs, although there have been some reports concerned with adverse effects. Such limited adverse effects observed in some trial studies include headache, inflammation and etc. additionally, considering to previous data, ADSC promote carcinoma progression and for that reason appear to increase the risk of cancer relapse in breast augmentation procedures. Therefore, there are needs for further research on understanding the potential application of ADSC as a safe and effective therapeutic option on diseases treatment in future.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Cell and tissue engineering

Country of origin: Iran

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Kolonin M, Tani K S- Editor: Kong JX L- Editor: A E- Editor: Lu YJ

References
1.  Marquez-Curtis LA, Janowska-Wieczorek A, McGann LE, Elliott JA. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects. Cryobiology. 2015;71:181-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 220]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
2.  Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279-4295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4817]  [Cited by in F6Publishing: 4858]  [Article Influence: 220.8]  [Reference Citation Analysis (0)]
3.  Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5703]  [Cited by in F6Publishing: 5560]  [Article Influence: 241.7]  [Reference Citation Analysis (0)]
4.  Sheykhhasan M, Qomi RT, Ghiasi M. Fibrin Scaffolds Designing in order to Human Adipose-derived Mesenchymal Stem Cells Differentiation to Chondrocytes in the Presence of TGF-β3. Int J Stem Cells. 2015;8:219-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
5.  Sheykhhasan M, Qomi RT, Kalhor N, Mehdizadeh M, Ghiasi M. Evaluation of the ability of natural and synthetic scaffolds in providing an appropriate environment for growth and chondrogenic differentiation of adipose-derived mesenchymal stem cells. Indian J Orthop. 2015;49:561-568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
6.  Zhao Q, Ren H, Han Z. Mesenchymal stem cells: Immunomodulatory capability and clinical potential in immune diseases. Journal of Cellular Immunotherapy. 2016;2:3-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 187]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
7.  Álvarez-Viejo M, Menéndez-Menéndez Y, Otero-Hernández J. CD271 as a marker to identify mesenchymal stem cells from diverse sources before culture. World J Stem Cells. 2015;7:470-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 99]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
8.  Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012;21:2724-2752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 540]  [Cited by in F6Publishing: 555]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
9.  Frese L, Dijkman PE, Hoerstrup SP. Adipose Tissue-Derived Stem Cells in Regenerative Medicine. Transfus Med Hemother. 2016;43:268-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 244]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
10.  Zack-Williams SD, Butler PE, Kalaskar DM. Current progress in use of adipose derived stem cells in peripheral nerve regeneration. World J Stem Cells. 2015;7:51-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
11.  Baer PC, Geiger H. Adipose-derived mesenchymal stromal/stem cells: tissue localization, characterization, and heterogeneity. Stem Cells Int. 2012;2012:812693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 309]  [Article Influence: 25.8]  [Reference Citation Analysis (0)]
12.  Fathi E, Farahzadi R. Isolation, Culturing, Characterization and Aging of Adipose Tissue-derived Mesenchymal Stem Cells: A Brief Overview. Braz Arch Biol Technol. 2016;59:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Ghiasi M, Tabatabaei Qomi M, Kalhor N, Qomi RT, Sheykhhasan M. The effects of synthetic and natural scaffolds on viability and proliferation of adipose-derived stem cells. Frontiers in Life Science. 2016;9:32-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
14.  Mundra V, Gerling IC, Mahato RI. Mesenchymal stem cell-based therapy. Mol Pharm. 2013;10:77-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
15.  Ryan JM, Barry FP, Murphy JM, Mahon BP. Mesenchymal stem cells avoid allogeneic rejection. J Inflamm (Lond). 2005;2:8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 598]  [Cited by in F6Publishing: 606]  [Article Influence: 31.9]  [Reference Citation Analysis (0)]
16.  Lin CS, Ning H, Lin G, Lue TF. Is CD34 truly a negative marker for mesenchymal stromal cells? Cytotherapy. 2012;14:1159-1163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 159]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
17.  Katz AJ, Tholpady A, Tholpady SS, Shang H, Ogle RC. Cell surface and transcriptional characterization of human adipose-derived adherent stromal (hADAS) cells. Stem Cells. 2005;23:412-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 381]  [Cited by in F6Publishing: 640]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
18.  Gronthos S, Franklin DM, Leddy HA, Robey PG, Storms RW, Gimble JM. Surface protein characterization of human adipose tissue-derived stromal cells. J Cell Physiol. 2001;189:54-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 780]  [Cited by in F6Publishing: 977]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
19.  Mitchell JB, McIntosh K, Zvonic S, Garrett S, Floyd ZE, Kloster A, Di Halvorsen Y, Storms RW, Goh B, Kilroy G. Immunophenotype of human adipose-derived cells: temporal changes in stromal-associated and stem cell-associated markers. Stem Cells. 2006;24:376-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 822]  [Cited by in F6Publishing: 1031]  [Article Influence: 54.3]  [Reference Citation Analysis (0)]
20.  Lindroos B, Aho KL, Kuokkanen H, Räty S, Huhtala H, Lemponen R, Yli-Harja O, Suuronen R, Miettinen S. Differential gene expression in adipose stem cells cultured in allogeneic human serum versus fetal bovine serum. Tissue Eng Part A. 2010;16:2281-2294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
21.  Zannettino AC, Paton S, Arthur A, Khor F, Itescu S, Gimble JM, Gronthos S. Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol. 2008;214:413-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 427]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
22.  Lindroos B, Boucher S, Chase L, Kuokkanen H, Huhtala H, Haataja R, Vemuri M, Suuronen R, Miettinen S. Serum-free, xeno-free culture media maintain the proliferation rate and multipotentiality of adipose stem cells in vitro. Cytotherapy. 2009;11:958-972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 156]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
23.  Parker A, Shang H, Khurgel M, Katz A. Low serum and serum-free culture of multipotential human adipose stem cells. Cytotherapy. 2007;9:637-646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 59]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
24.  Mirabet V, Solves P, Miñana MD, Encabo A, Carbonell-Uberos F, Blanquer A, Roig R. Human platelet lysate enhances the proliferative activity of cultured human fibroblast-like cells from different tissues. Cell Tissue Bank. 2008;9:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
25.  Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24:1294-1301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2248]  [Cited by in F6Publishing: 2215]  [Article Influence: 123.1]  [Reference Citation Analysis (0)]
26.  Liu S, Zhou J, Zhang X, Liu Y, Chen J, Hu B, Song J, Zhang Y. Strategies to Optimize Adult Stem Cell Therapy for Tissue Regeneration. Int J Mol Sci. 2016;17:982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 93]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
27.  Zhu P, Liu J, Shi J, Zhou Q, Liu J, Zhang X, Du Z, Liu Q, Guo Y. Melatonin protects ADSCs from ROS and enhances their therapeutic potency in a rat model of myocardial infarction. J Cell Mol Med. 2015;19:2232-2243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
28.  Tan SS, Han X, Sivakumaran P, Lim SY, Morrison WA. Melatonin Protects Human Adipose-Derived Stem Cells from Oxidative Stress and Cell Death. Arch Plast Surg. 2016;43:237-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
29.  Bajek A, Gurtowska N, Olkowska J, Kazmierski L, Maj M, Drewa T. Adipose-Derived Stem Cells as a Tool in Cell-Based Therapies. Arch Immunol Ther Exp (Warsz). 2016;64:443-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 119]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
30.  Search for Studies [Internet] Home - ClinicalTrials.gov. [accessed. 2017;Jan 25] Available from: https://clinicaltrials.gov/.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Pak J, Lee JH, Park KS, Park M, Kang LW, Lee SH. Current use of autologous adipose tissue-derived stromal vascular fraction cells for orthopedic applications. J Biomed Sci. 2017;24:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 63]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
32.  Pak J, Chang JJ, Lee JH, Lee SH. Safety reporting on implantation of autologous adipose tissue-derived stem cells with platelet-rich plasma into human articular joints. BMC Musculoskelet Disord. 2013;14:337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 113]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
33.  Perdisa F, Gostyńska N, Roffi A, Filardo G, Marcacci M, Kon E. Adipose-Derived Mesenchymal Stem Cells for the Treatment of Articular Cartilage: A Systematic Review on Preclinical and Clinical Evidence. Stem Cells Int. 2015;2015:597652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 75]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
34.  Berman M, Lander E. A Prospective Safety Study of Autologous Adipose-Derived Stromal Vascular Fraction Using a Specialized Surgical Processing System. American J of Cosmetic Surg. 2017;1-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
35.  Staff NP, Madigan NN, Morris J, Jentoft M, Sorenson EJ, Butler G, Gastineau D, Dietz A, Windebank AJ. Safety of intrathecal autologous adipose-derived mesenchymal stromal cells in patients with ALS. Neurology. 2016;87:2230-2234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 76]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
36.  Lander EB, Berman MH, See JR. Stromal Vascular Fraction Combined with Shock Wave for the Treatment of Peyronie’s Disease. Plast Reconstr Surg Glob Open. 2016;4:e631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
37.  Minteer DM, Marra KG, Rubin JP. Adipose stem cells: biology, safety, regulation, and regenerative potential. Clin Plast Surg. 2015;42:169-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
38.  Fodor PB, Paulseth SG. Adipose Derived Stromal Cell (ADSC) Injections for Pain Management of Osteoarthritis in the Human Knee Joint. Aesthet Surg J. 2016;36:229-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 98]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
39.  Sanz-Baro R, García-Arranz M, Guadalajara H, de la Quintana P, Herreros MD, García-Olmo D. First-in-Human Case Study: Pregnancy in Women With Crohn’s Perianal Fistula Treated With Adipose-Derived Stem Cells: A Safety Study. Stem Cells Transl Med. 2015;4:598-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
40.  Grønhøj C, Jensen DH, Glovinski PV, Jensen SB, Bardow A, Oliveri RS, Specht L, Thomsen C, Darkner S, Kiss K. First-in-man mesenchymal stem cells for radiation-induced xerostomia (MESRIX): study protocol for a randomized controlled trial. Trials. 2017;18:108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
41.  Le PTB, Pham PV, Dang LTT, Phan NK. Expanded autologous adipose derived stem cell transplantation for type 2 diabetes mellitus. Biomed Res Ther. 2016;3:1034-1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
42.  Vu NB, Trinh NV, Phi LT, Vo TLH, Dao TTT, Phan NK, Van Ta T, Van Pham P. An evaluation of the safety of adipose-derived stem cells. Biomed Res Ther. 2015;2:359-365.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Boennelycke M, Gras S, Lose G. Tissue engineering as a potential alternative or adjunct to surgical reconstruction in treating pelvic organ prolapse. Int Urogynecol J. 2013;24:741-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
44.  Comella K, Silbert R, Parlo M. Effects of the intradiscal implantation of stromal vascular fraction plus platelet rich plasma in patients with degenerative disc disease. J Transl Med. 2017;15:12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 58]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
45.  Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, Kim JE, Shim H, Shin JS, Shin IS. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells. 2014;32:1254-1266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 566]  [Cited by in F6Publishing: 584]  [Article Influence: 58.4]  [Reference Citation Analysis (0)]
46.  Pham PV. Adipose stem cells in the clinic. Biomed Res Ther. 2014;1:57-70 [DO I: 10.7603/s40730-014-0011-8].  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Wei HJ, Zeng R, Lu JH, Lai WF, Chen WH, Liu HY, Chang YT, Deng WP. Adipose-derived stem cells promote tumor initiation and accelerate tumor growth by interleukin-6 production. Oncotarget. 2015;6:7713-7726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
48.  Schweizer R, Tsuji W, Gorantla VS, Marra KG, Rubin JP, Plock JA. The role of adipose-derived stem cells in breast cancer progression and metastasis. Stem Cells Int. 2015;2015:120949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
49.  Guillaume-Jugnot P, Daumas A, Magalon J, Jouve E, Nguyen PS, Truillet R, Mallet S, Casanova D, Giraudo L, Veran J. Autologous adipose-derived stromal vascular fraction in patients with systemic sclerosis: 12-month follow-up. Rheumatology (Oxford). 2016;55:301-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
50.  Kim YS, Lee HJ, Choi YJ, Kim YI, Koh YG. Does an injection of a stromal vascular fraction containing adipose-derived mesenchymal stem cells influence the outcomes of marrow stimulation in osteochondral lesions of the talus? A clinical and magnetic resonance imaging study. Am J Sports Med. 2014;42:2424-2434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 64]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
51.  Anderi R, Makdissy N, Rizk F, Hamade A. Hair quality improvement in alopecia patients following adipose-derived stem cell treatment. JPRAS Open. 2017;.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Shin H, Ryu HH, Kwon O, Park BS, Jo SJ. Clinical use of conditioned media of adipose tissue-derived stem cells in female pattern hair loss: a retrospective case series study. Int J Dermatol. 2015;54:730-735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 81]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
53.  Fukuoka H, Suga H. Hair Regeneration Treatment Using Adipose-Derived Stem Cell Conditioned Medium: Follow-up With Trichograms. Eplasty. 2015;15:e10.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Granel B, Daumas A, Jouve E, Harlé JR, Nguyen PS, Chabannon C, Colavolpe N, Reynier JC, Truillet R, Mallet S. Safety, tolerability and potential efficacy of injection of autologous adipose-derived stromal vascular fraction in the fingers of patients with systemic sclerosis: an open-label phase I trial. Ann Rheum Dis. 2015;74:2175-2182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 122]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
55.  Daumas A, Magalon J, Jouve E, Truillet R, Casanova D, Giraudo L, Veran J, Benyamine A, Dignat-George F, Magalon G. Long-term follow-up after autologous adipose-derived stromal vascular fraction injection into fingers in systemic sclerosis patients. Curr Res Transl Med. 2017;65:40-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 44]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
56.  Dave M, Jaiswal P, Cominelli F. Mesenchymal stem/stromal cell therapy for inflammatory bowel disease: an updated review with maintenance of remission. Curr Opin Gastroenterol. 2017;33:59-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
57.  González MA, Gonzalez-Rey E, Rico L, Büscher D, Delgado M. Adipose-derived mesenchymal stem cells alleviate experimental colitis by inhibiting inflammatory and autoimmune responses. Gastroenterology. 2009;136:978-989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 466]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
58.  Anderson P, Souza-Moreira L, Morell M, Caro M, O’Valle F, Gonzalez-Rey E, Delgado M. Adipose-derived mesenchymal stromal cells induce immunomodulatory macrophages which protect from experimental colitis and sepsis. Gut. 2013;62:1131-1141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 163]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
59.  Jung WY, Kang JH, Kim KG, Kim HS, Jang BI, Park YH, Song IH. Human adipose-derived stem cells attenuate inflammatory bowel disease in IL-10 knockout mice. Tissue Cell. 2015;47:86-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
60.  Liu WH, Song FQ, Ren LN, Guo WQ, Wang T, Feng YX, Tang LJ, Li K. The multiple functional roles of mesenchymal stem cells in participating in treating liver diseases. J Cell Mol Med. 2015;19:511-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 95]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
61.  de Girolamo L, Lucarelli E, Alessandri G, Avanzini MA, Bernardo ME, Biagi E, Brini AT, D’Amico G, Fagioli F, Ferrero I. Mesenchymal stem/stromal cells: a new ‘’cells as drugs’’ paradigm. Efficacy and critical aspects in cell therapy. Curr Pharm Des. 2013;19:2459-2473.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Zuk P. Adipose-Derived Stem Cells in Tissue Regeneration: A Review. ISRN Stem Cells. 2013;2013:1-35.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Cantz T, Sharma AD, Ott M. Concise review: cell therapies for hereditary metabolic liver diseases-concepts, clinical results, and future developments. Stem Cells. 2015;33:1055-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
64.  Miki T, Grubbs B. Therapeutic potential of placenta-derived stem cells for liver diseases: current status and perspectives. J Obstet Gynaecol Res. 2014;40:360-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
65.  Sun K, Xie X, Xie J, Jiao S, Chen X, Zhao X, Wang X, Wei L. Cell-based therapy for acute and chronic liver failures: distinct diseases, different choices. Sci Rep. 2014;4:6494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
66.  Chen G, Jin Y, Shi X, Qiu Y, Zhang Y, Cheng M, Wang X, Chen C, Wu Y, Jiang F. Adipose-derived stem cell-based treatment for acute liver failure. Stem Cell Res Ther. 2015;6:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
67.  Hu C, Li L. In vitro and In vivo Hepatic Differentiation of Adult Somatic Stem Cells and Extraembryonic Stem Cells for Treating End Stage Liver Diseases. Stem Cells Int. 2015;2015:871972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
68.  Nguyen A, Guo J, Banyard DA, Fadavi D, Toranto JD, Wirth GA, Paydar KZ, Evans GR, Widgerow AD. Stromal vascular fraction: A regenerative reality? Part 1: Current concepts and review of the literature. J Plast Reconstr Aesthet Surg. 2016;69:170-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 89]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
69.  Rajashekhar G, Ramadan A, Abburi C, Callaghan B, Traktuev DO, Evans-Molina C, Maturi R, Harris A, Kern TS, March KL. Regenerative therapeutic potential of adipose stromal cells in early stage diabetic retinopathy. PLoS One. 2014;9:e84671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 77]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
70.  Pacheco R, Contreras F, Zouali M. The dopaminergic system in autoimmune diseases. Front Immunol. 2014;5:117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 108]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
71.  Wankhade UD, Shen M, Kolhe R, Fulzele S. Advances in Adipose-Derived Stem Cells Isolation, Characterization, and Application in Regenerative Tissue Engineering. Stem Cells Int. 2016;2016:3206807.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Feisst V, Meidinger S, Locke MB. From bench to bedside: use of human adipose-derived stem cells. Stem Cells Cloning. 2015;8:149-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
73.  Riordan NH, Ichim TE, Min WP, Wang H, Solano F, Lara F, Alfaro M, Rodriguez JP, Harman RJ, Patel AN. Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis. J Transl Med. 2009;7:29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 234]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
74.  Bowles AC, Strong AL, Wise RM, Thomas RC, Gerstein BY, Dutreil MF, Hunter RS, Gimble JM, Bunnell BA. Adipose Stromal Vascular Fraction-Mediated Improvements at Late-Stage Disease in a Murine Model of Multiple Sclerosis. Stem Cells. 2017;35:532-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
75.  Stepien A, Dabrowska NL, Maciagowska M, Macoch RP, Zolocinska A, Mazur S, Siennicka K, Frankowska E, Kidzinski R, Chalimoniuk M. Clinical Application of Autologous Adipose Stem Cells in Patients with Multiple Sclerosis: Preliminary Results. Mediators Inflamm. 2016;2016:5302120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 30]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
76.  Hedayatpour A, Ragerdi I, Pasbakhsh P, Kafami L, Atlasi N, Pirhajati Mahabadi V, Ghasemi S, Reza M. Promotion of remyelination by adipose mesenchymal stem cell transplantation in a cuprizone model of multiple sclerosis. Cell J. 2013;15:142-151.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Shalaby SM, Sabbah NA, Saber T, Abdel Hamid RA. Adipose-derived mesenchymal stem cells modulate the immune response in chronic experimental autoimmune encephalomyelitis model. IUBMB Life. 2016;68:106-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
78.  Strong AL, Bowles AC, Wise RM, Morand JP, Dutreil MF, Gimble JM, Bunnell BA. Human Adipose Stromal/Stem Cells from Obese Donors Show Reduced Efficacy in Halting Disease Progression in the Experimental Autoimmune Encephalomyelitis Model of Multiple Sclerosis. Stem Cells. 2016;34:614-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
79.  Yousefi F, Ebtekar M, Soudi S, Soleimani M, Hashemi SM. In vivo immunomodulatory effects of adipose-derived mesenchymal stem cells conditioned medium in experimental autoimmune encephalomyelitis. Immunol Lett. 2016;172:94-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
80.  Onesti MG, Fioramonti P, Carella S, Fino P, Marchese C, Scuderi N. Improvement of Mouth Functional Disability in Systemic Sclerosis Patients over One Year in a Trial of Fat Transplantation versus Adipose-Derived Stromal Cells. Stem Cells Int. 2016;2016:2416192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
81.  Freitag J, Bates D, Boyd R, Shah K, Barnard A, Huguenin L, Tenen A. Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy - a review. BMC Musculoskelet Disord. 2016;17:230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 172]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
82.  Tantuway V, Bhambani P, Nagla A, Mantry P, Sharma R, Mehto P, Bansal S. Autologous grafting of non manupulated freshly isolated - adipose tissue derived stromal vascular fraction in single surgical sitting for treatment of knee osteoarthritis. International Journal of Research in Orthopaedics. 2016;3:107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
83.  Pérez-Silos V, Camacho-Morales A, Fuentes-Mera L. Mesenchymal Stem Cells Subpopulations: Application for Orthopedic Regenerative Medicine. Stem Cells Int. 2016;2016:3187491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
84.  Nguyen PD, Tran TD, Nguyen HT, Vu HT, Le PT, Phan NL, Vu NB, Phan NK, Van Pham P. Comparative Clinical Observation of Arthroscopic Microfracture in the Presence and Absence of a Stromal Vascular Fraction Injection for Osteoarthritis. Stem Cells Transl Med. 2017;6:187-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 65]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
85.  Pak J, Lee JH, Park KS, Jeong BC, Lee SH. Regeneration of Cartilage in Human Knee Osteoarthritis with Autologous Adipose Tissue-Derived Stem Cells and Autologous Extracellular Matrix. Biores Open Access. 2016;5:192-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 44]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
86.  Pak J, Lee JH, Kartolo WA, Lee SH. Cartilage Regeneration in Human with Adipose Tissue-Derived Stem Cells: Current Status in Clinical Implications. Biomed Res Int. 2016;2016:4702674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
87.  de Girolamo L, Arrigoni E, Stanco D, Lopa S, Di Giancamillo A, Addis A, Borgonovo S, Dellavia C, Domeneghini C, Brini AT. Role of autologous rabbit adipose-derived stem cells in the early phases of the repairing process of critical bone defects. J Orthop Res. 2011;29:100-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
88.  Bhattacharya I, Ghayor C, Weber FE. The Use of Adipose Tissue-Derived Progenitors in Bone Tissue Engineering - a Review. Transfus Med Hemother. 2016;43:336-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
89.  Im GI. Regeneration of articular cartilage using adipose stem cells. J Biomed Mater Res A. 2016;104:1830-1844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
90.  Yang Y, Choi H, Seon M, Cho D, Bang SI. LL-37 stimulates the functions of adipose-derived stromal/stem cells via early growth response 1 and the MAPK pathway. Stem Cell Res Ther. 2016;7:58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
91.  Ramdasi S, Tiwari SK. Human Mesenchymal Stem Cell-Derived Conditioned Media for Hair Regeneration Applications. J Stem Cells. 2016;11:201-211.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Jin SE, Sung JH. Hair regeneration using adipose-derived stem cells. Histol Histopathol. 2016;31:249-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
93.  Park BS, Kim WS. Adipose-Derived Stem Cells and Their Secretory Factors for Skin Aging and Hair Loss. Textbook of Aging Skin. Berlin Heidelberg: Springer Press 2017; 205-224.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Fukuoka H, Suga H, Narita K, Watanabe R, Shintani S. The latest advance in hair regeneration therapy using proteins secreted by adipose-derived stem cells. Am J Cosmet Surg. 2012;29:273-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
95.  Moon KM, Park YH, Lee JS, Chae YB, Kim MM, Kim DS, Kim BW, Nam SW, Lee JH. The effect of secretory factors of adipose-derived stem cells on human keratinocytes. Int J Mol Sci. 2012;13:1239-1257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 82]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
96.  Park BS, Kim WS, Choi JS, Kim HK, Won JH, Ohkubo F, Fukuoka H. Hair growth stimulated by conditioned medium of adipose-derived stem cells is enhanced by hypoxia: evidence of increased growth factor secretion. Biomed Res. 2010;31:27-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 116]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
97.  Huang CF, Chang YJ, Hsueh YY, Huang CW, Wang DH, Huang TC, Wu YT, Su FC, Hughes M, Chuong CM. Assembling Composite Dermal Papilla Spheres with Adipose-derived Stem Cells to Enhance Hair Follicle Induction. Sci Rep. 2016;6:26436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
98.  Festa E, Fretz J, Berry R, Schmidt B, Rodeheffer M, Horowitz M, Horsley V. Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell. 2011;146:761-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 413]  [Cited by in F6Publishing: 422]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
99.  Gaur M, Dobke M, Lunyak VV. Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging. Int J Mol Sci. 2017;18:208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 105]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
100.  Lee A, Bae S, Lee SH, Kweon OK, Kim WH. Hair growth promoting effect of dermal papilla like tissues from canine adipose-derived mesenchymal stem cells through vascular endothelial growth factor. J Vet Med Sci. 2017;78:1811-1818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
101.  Yoshida A, Kitajiri S, Nakagawa T, Hashido K, Inaoka T, Ito J. Adipose tissue-derived stromal cells protect hair cells from aminoglycoside. Laryngoscope. 2011;121:1281-1286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
102.  Fouad H, Sabry D, Elsetohy K, Fathy N. Therapeutic efficacy of amniotic membrane stem cells and adipose tissue stem cells in rats with chemically induced ovarian failure. J Adv Res. 2016;7:233-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
103.  Omar FR, Amin NMA, Elsherif HA, Mohamed DH. Role of Adipose-Derived Stem Cells in Restoring Ovarian Structure of Adult Albino Rats with Chemotherapy-Induced Ovarian Failure: A Histological and Immunohistochemical Study. J Carcinog Mutagen. 2016;7:254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
104.  Su J, Ding L, Cheng J, Yang J, Li X, Yan G, Sun H, Dai J, Hu Y. Transplantation of adipose-derived stem cells combined with collagen scaffolds restores ovarian function in a rat model of premature ovarian insufficiency. Hum Reprod. 2016;31:1075-1086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 86]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
105.  Sun M, Wang S, Li Y, Yu L, Gu F, Wang C, Yao Y. Adipose-derived stem cells improved mouse ovary function after chemotherapy-induced ovary failure. Stem Cell Res Ther. 2013;4:80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 123]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
106.  Ghiasi M, Fazaely H, Asaii E, Sheykhhasan M. In vitro Maturation of Human Oocytes using Conditioned Medium of Mesenchymal Stem Cells and Formation of Embryo by Use of ICSI. SMU Medical Journal. 2014;1:89-98.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Cakici C, Buyrukcu B, Duruksu G, Haliloglu AH, Aksoy A, Isýk A, Uludag O, Ustun H, Subasý C, Karaoz E. Recovery of fertility in azoospermia rats after injection of adipose-tissue-derived mesenchymal stem cells: the sperm generation. Biomed Res Int. 2013;2013:529589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 72]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
108.  Fazaeli H, Faeze D, Naser K, Maryam S, Mohammad M, Mahdieh G, TQ Reza. Introducing of a New Experimental Method in Semen Preparation: Supernatant Product of Adipose Tissue: Derived Mesenchymal Stem Cells (SPAS). The Patient’s perspective. JFIV Reprod Med Genet. 2016;4:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
109.  Terraciano P, Garcez T, Ayres L, Durli I, Baggio M, Kuhl CP, Laurino C, Passos E, Paz AH, Cirne-Lima E. Cell therapy for chemically induced ovarian failure in mice. Stem Cells Int. 2014;2014:720753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 40]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
110.  Gokce A, Abd Elmageed ZY, Lasker GF, Bouljihad M, Kim H, Trost LW, Kadowitz PJ, Abdel-Mageed AB, Sikka SC, Hellstrom WJ. Adipose tissue-derived stem cell therapy for prevention and treatment of erectile dysfunction in a rat model of Peyronie’s disease. Andrology. 2014;2:244-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 61]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
111.  Castiglione F, Hedlund P, Van der Aa F, Bivalacqua TJ, Rigatti P, Van Poppel H, Montorsi F, De Ridder D, Albersen M. Intratunical injection of human adipose tissue-derived stem cells prevents fibrosis and is associated with improved erectile function in a rat model of Peyronie’s disease. Eur Urol. 2013;63:551-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 127]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
112.  Piao S, Ryu JK, Shin HY, Zhang L, Song SU, Han JY, Park SH, Kim JM, Kim IH, Kim SJ. Repeated intratunical injection of adenovirus expressing transforming growth factor-beta1 in a rat induces penile curvature with tunical fibrotic plaque: a useful model for the study of Peyronie’s disease. Int J Androl. 2008;31:346-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
113.  Sangkum P. Research highlights on stem cell therapy for the treatment of Peyronie’s disease. Transl Androl Urol. 2016;5:363-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
114.  Lim JY, Ra JC, Shin IS, Jang YH, An HY, Choi JS, Kim WC, Kim YM. Systemic transplantation of human adipose tissue-derived mesenchymal stem cells for the regeneration of irradiation-induced salivary gland damage. PLoS One. 2013;8:e71167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 92]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
115.  Li Z, Wang Y, Xing H, Wang Z, Hu H, An R, Xu H, Liu Y, Liu B. Protective efficacy of intravenous transplantation of adipose-derived stem cells for the prevention of radiation-induced salivary gland damage. Arch Oral Biol. 2015;60:1488-1496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
116.  Kojima T, Kanemaru S, Hirano S, Tateya I, Ohno S, Nakamura T, Ito J. Regeneration of radiation damaged salivary glands with adipose-derived stromal cells. Laryngoscope. 2011;121:1864-1869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
117.  An HY, Shin HS, Choi JS, Kim HJ, Lim JY, Kim YM. Adipose Mesenchymal Stem Cell Secretome Modulated in Hypoxia for Remodeling of Radiation-Induced Salivary Gland Damage. PLoS One. 2015;10:e0141862.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
118.  Choi JS, An HY, Shin HS, Kim YM, Lim JY. Enhanced tissue remodelling efficacy of adipose-derived mesenchymal stem cells using injectable matrices in radiation-damaged salivary gland model. J Tissue Eng Regen Med. 2016; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
119.  Maria OM, Tran SD. Human mesenchymal stem cells cultured with salivary gland biopsies adopt an epithelial phenotype. Stem Cells Dev. 2011;20:959-967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
120.  Lombaert I, Movahednia MM, Adine C, Ferreira JN. Concise Review: Salivary Gland Regeneration: Therapeutic Approaches from Stem Cells to Tissue Organoids. Stem Cells. 2017;35:97-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 89]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
121.  Yoo C, Vines JB, Alexander G, Murdock K, Hwang P, Jun HW. Adult stem cells and tissue engineering strategies for salivary gland regeneration: a review. Biomater Res. 2014;18:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
122.  Xiong X, Shi X, Chen F. Human adipose tissuederived stem cells alleviate radiationinduced xerostomia. Int J Mol Med. 2014;34:749-755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
123.  Kawakami M, Ishikawa H, Tanaka A, Mataga I. Induction and differentiation of adipose-derived stem cells from human buccal fat pads into salivary gland cells. Hum Cell. 2016;29:101-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
124.  Lee J, Park S, Roh S. Transdifferentiation of mouse adipose-derived stromal cells into acinar cells of the submandibular gland using a co-culture system. Exp Cell Res. 2015;334:160-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]