banner



Is It Necessary To Grow Human Skin On A Rat For Burn Victims?

i. INTRODUCTION

Despite the known take chances of mortality associated with severe burn injury, improved resuscitation and handling of inhalation injury, advisable infection control, and standardized critical care accept improved the clinical issue of severely burned patients in recent years. However, severe fire remains a devastating injury affecting nearly every organ and leading to significant morbidity and mortality.

Stalk cell therapy has recently emerged as an innovative treatment for fire injuries, hypertrophic scar, or sepsis.1–3 Literature survey reveals that mesenchymal stem cells (MSCs) contribute not merely to local wound healing but also participate in systemic inflammation responses. The severe burn-induced persistent inflammatory response increases susceptibility to infections and sepsis, potentially leading to multiorgan failure and decease. During major burn injury-induced inflammatory response, proinflammatory cytokines such as interleukin (IL)-1β, IL-half dozen, IL-viii, tumor necrosis gene (TNF)-α or interferon (IFN)-γ, and anti-inflammatory cytokines such as IL-4, IL-10, and granulocyte-colony stimulating factor are released.4–six Several studies accept reported that the levels of various cytokines are loftier in burn patients; increased cytokine levels are also related to sepsis and mortality in these patients.half-dozen MSCs are known to part at several levels of the inflammatory response, specially in the early phase of sepsis, to regulate a broad console of inflammatory cytokines and inhibit leukocyte infiltration into several target organs.7 , 8 MSC likewise contribute to skin appendage regeneration and wound healing through MAPK/ERK signal pathway.9 Significant evidences from basic and clinical research supporting MSCs angiogenic role seen in the handling of ischemic limbs, myocardial infarction, and retinopathy are emerging.10–12 Recently, the utilize of adipose-derived stem cells (ASCs) has emerged in cell therapy. ASCs have the advantage of being easily and abundantly available from fat tissue extracted by liposuction. ASCs can differentiate into unlike lineages and stimulate the release of multiple growth factors.13 Their therapeutic furnishings are being investigated for weather such as degenerative articulatio genus, renal failure, hepatic failure, and myocardial infarction.14–17 ASCs accept institute application in the engineering of various kinds of tissue, such as bone, auricular tissue, periodontal tissue, and tendon.18–21

All the same, in that location are few studies reporting the application of ASCs in treating acute burn injuries and its effectiveness is yet uncertain. In a systematic review, Conde´-Green et al revealed that fat grafts and ASCs have benign effects on acute burn wound healing, but also pointed out the lack of decisive prove.22 Using a mouse model, Bliley et al showed that treatment with human ASCs improved healing of full-thickness fire wound observed as a reduction in wound surface area; ASCs handling improved wound healing past enhancing vascularity, collagen degradation, and adipogenesis.3 Withal, the effect of ASCs on peel appendage regeneration in burn injury has not been studied. Furthermore, efficient and cost-constructive treatments that can advance burn down wound healing, decrease the size of burn wound excision area, and reduce the tissue necrosis past preventing conversion of deep partial thickness to full-thickness burns do not exist.

Thus, the aim of our study is to investigate the upshot of locally administered ASCs on deep partial thickness burn wound healing in rat. To convalesce interference from main wound contraction in rodents, which is limited in the humans simply could derange quantitative and qualitative evaluation of wound repair in these animal models,23 we designed an innovative skin island burn down wound model in rat and farther used it to evaluate the wound healing and skin appendage regeneration after local injection of ASCs.

2. METHODS

two.ane. Cell isolation and culture

Rat ASCs were derived from inguinal fat pad, which was isolated and cultured from Sprague Dawley (SD) rat (250–300 chiliad, BioLASCO Taiwan Co., Ltd) as previously described.fifteen , nineteen The cell surface markers CD11B, CD29, CD31, CD45, and CD90 were analyzed by menses-cytometry (FACS Canto II; Becton Dickinson, Franklin Lakes, NJ, USA). The differentiation of ASCs into bone and adipose in the presence of the induction mediumfifteen was checked with von Kossa/Oil red O staining (Sigma-Aldrich, Taiwan) under light microscope (Olympus IX51, OLYMPUS Corporation, Tokyo, Japan). ASCs from passages three to six were used for local injection.

2.two. Skin isle burn wound rat model

The study was approved past the Animal Care/Utilise Committee of Taipei Veterans General Infirmary (IACUC2014-163). We created a contact burn down injury model in SD rats. For this, the SD rat was anesthetized with Zoletil (0.1 mL/100 mg) by intraperitoneal injection. Fig. 1 illustrates the model nonlethal contact burn wound on the pare island on the dorsum of the rat. A copper plate, 1 cm × 1 cm in size, heated to 90°C (by placing in humid h2o), was applied for 30 s over the assigned depilated peel area on the rat'due south back. Each skin isle had three such contact burn wounds, surface area of each of these wound units corresponded to the area of copper plate (ane cm × 1 cm). Post injury and after awarding of corresponding treatments the skin islands were embedded into the subcutaneous pockets by suturing the skin around circumferential incision, Fig. 1D. This model was used for the following two reasons—(1) reduction in number of experimental animals used and (2) interference by wound wrinkle and epithelium migration from the side by side uninjured skin could be avoided past isolating the injured skin from adjacent skin by the circumferential incision.

F1-8
Fig. 1:

A, The assignment and treatment protocols for nonlethal contact burn down wounds on ii strips of peel islands on the depilated rat back; each skin island had 3 burn wounds induced past 30 due south awarding of heated copper plate, with each wound area corresponding to copper plate area (1 cm × 1 cm); B, Adipose-derived stem cells (ASCs)-treated group—three wounds in a peel isle was locally intradermally injected with 5 × 105 ASCs; Control group—the three wounds in another skin island in the same animal were injected with PBS; Progress in healing was checked regularly at four different fourth dimension-points; experiments were carried out in triplicates, ie, observations were carried out on three rats (total n = 9 for each grouping); C, After injection, the skin around the pare island was incised circumferentially to avoid wound wrinkle; D, Finally, the pare around the pare island was sutured with nylon thread thus in issue embedding the pare island in subcutaneous pocket.

2.three. Localized injection of ASCs into the rat fire wound

To evaluate the outcome of locally injected ASCs on the healing of burn wounds, progress in healing were compared between control wounds and ASCs-treated fire wounds in the same fauna. Two pare islands, each had iii contact burn wounds, were designed and divided into two groups—Control group (north = 3) and ASCs-treated (due north = three) wound groups (Fig. 1A). Thirty minutes later on the awarding of contact fire protocol, the wounds in control group were intradermally injected with 0.2 mL of PBS, and the wounds in ASCs-treated group received intradermal injections of v × 105 ASCs resuspended in 0.ii mL of PBS. Progress in wound healing was checked at different time intervals after treatment, ie, at 1st, 2d, 3rd, and 4th weeks (three rats at each time-point; full 12 rats); the sutured skin isle was opened and its gross appearance was imaged using a standardized digital photograph. The mean wound expanse was calculated using ImageJ (National Institutes of Health, Bethesda, Medico, U.s.a.). At each fourth dimension-point, nine whole-layer skin samples of contact burn wounds per group were obtained for histological (H&E) and immunocytochemical studies (CD31 and PNCA staining; Abcam). The total number of follicles and live follicles were measured under ×200 magnification. Vascular density and expression of proliferating cell nuclear antigen (PCNA) were evaluated.

2.4. Statistical analysis

The grouping differences were analyzed in SPSS 12.0 past using two-tailed paired Educatee's t test and 1-way analysis of variance. The quantitative results are presented as mean ± SD. p value <0.05 indicated statistical significance.

3. RESULTS

3.1. Label and differentiation of rat ASCs

Harvested rat ASCs was passaged thrice and earlier being injecting into the rats, they were subjected to menstruum cytometric analysis and differentiation assays. We found that purified rat ASCs expressed high levels of CD29 and CD90, whereas CD11B, CD31, and CD45 (Fig. 2A) were not highly expressed. After induction of differentiation, rat ASCs were found to differentiate via adipogenesis and osteogenesis (Fig. 2B).

F2-8
Fig. 2:

A, The adipose-derived stem cells (ASCs) characterized by flow cytometry were positive for CD29 and CD90 surface staining (to a higher place) just non for CD11B, CD31, and CD45 surface staining (below); B, When induced, the ASCs exhibited adipogenic and osteogenic differentiation; Adipogenic differentiation forming lipid vesicles was confirmed with oil red O staining (left); Calcium deposits stained with von Kossa confirmed osteogenesis (right).

3.2. Confirmation of burn injury

In this study, in each of the 12 animals used for the experiments, a total of 6 deep partial thickness burn wounds were induced and were divided every bit into control and ASCs-treated groups (ie, each rat had three command burn wounds and iii ASCs-treated burn down wounds); and observations on recovery were made at four different time-points (on 1st, 2nd, third, and 4th week). During the entire period of the experiments, no wound infections or systemic adverse events were observed in any of the experimental animals. The depth of burn injury was confirmed by histopathological studies, which showed that necrotic tissue extended to reticular dermis indicating that partial-thickness burns were indeed induced by the protocol used in this report (Fig. iii).

F3-8
Fig. iii:

The depth of fire injury was confirmed past histopathological studies, which showed that necrotic levels involved the reticular dermis (arrows), indicating deep fractional thickness burn wound.

iii.3. Wound healing and skin appendage regeneration

To chart the grade of burn wound healing, the sutured skin was opened at different intervals of time (at 1st, 2nd, 3rd, and 4th week) and gross appearance of the embedded pare islands was recorded. For comparison, the mean areas of the deep partial thickness contact burn down wounds at iv unlike fourth dimension-points were measured. It was constitute that at all given fourth dimension points the wound healing in ASCs-treated groups was improved as indicated by improved hair growth in ASCs-treated wounds as compared with control groups. This finding was truthful peculiarly in the first 2 weeks (p < 0.05) (Fig. 4A, B). To evaluate the effect of ASCs on wound healing, the follicular density was measured under ×200 magnification. It was found that betwixt the 3rd and fourth weeks, the pct of live follicles increased gradually in the ASCs-treated groups compared with control groups (p < 0.05) (Fig. 4C).

F4-8
Fig. 4:

A, As early on equally the offset week subsequently burn down injury, wound healing was improved in adipose-derived stalk cells (ASCs)-treated group with more hair growth when compared to control grouping; B, A greater decrease in the hateful wound area of deep partial thickness fire wounds and better healing was observed in ASCs-treated group when compared to control grouping, specially in the first ii weeks afterward injury; C, The per centum of live follicles, observed under low-cal microscope, increased gradually in ASCs-treated group, uniform with gross findings (statistical significance is indicated as follows: *p < 0.05; **p < 0.01; ***p < 0.001).

three.4. Mechanism of ASCs-mediated fire wound healing

The mechanism of ASCs-mediated wound healing was likewise investigated through CD31 and PCNA staining of the skin biopsy samples. Angiogenesis was observed in ASCs-treated group during the third and 4th weeks mail injury as indicated by the significantly higher number of CD31 positive cells and increased vascular density every bit compared with the control group (p < 0.001). Meaning increase in vascular density was observed in ASCs-treated groups beginning from 1st week of recovery (Fig. 5A). The nucleus of stromal cells including endothelial cells and fibroblasts were PCNA positive; PCNA positive cells were counted to estimate PCNA index. PCNA index was significantly increased by the 3rd week in the ASCs-treated group compared with control group (p < 0.05) (Fig. 5B).

F5-8
Fig. 5:

A, Local injection of adipose-derived stem cells (ASCs) increases vascular density and angiogenesis in deep fractional thickness contact burn wound; B, Proliferating cell nuclear antigen (PCNA) index showed significant increase in the 3rd calendar week in the ASCs-treated group compared with command group (statistical significance is indicated equally follows: *p < 0.05; **p < 0.01; ***p < 0.001).

4. DISCUSSION

In severe fire injuries, burn patients suffer from not only massive skin loss but also excessive systemic inflammatory responses. Delay in wound healing enhances risks of burn wound sepsis or infection; uninterrupted savage inflammatory bicycle increases bloodshed. Although considerable progress has been accomplished in burn treatment techniques such as synthetic dressing, debridement and skin grafting techniques, tissue-engineered peel substitutes, and topical growth factors application, methods to accordingly regenerate skin appendages and effectively prevent hypertrophic scarring are defective.

Our skin isle burn wound model was designed to alleviate interference from wound contraction in rodents. In previously studied models, mechanical fixation devices or splints have been employed to avoid wound contraction.24 , 25 Although these methods were useful in keeping the wound volume relatively constant and thus allowing morphological or biomolecular quantification of wound response, many limitations remain. Using such mechanical devices is very complicated and due consideration must be given to application process and durability of the application. Moreover, these mechanical procedures are limited in stopping the normal influx of local cells to the wound site.25 The skin isle method designed here was plant to be convenient and easily implementable. Furthermore, the circumferential incision prevented migration of cells and appendages from next good for you skin and also protected the wounds in the island from infection. This skin island wound model method for evaluating wound healing is novel and not mentioned in literature to date.

This study demonstrated that local injection of ASCs into burn down wounds tin can be performed with ease in rat models; ASCs injection promoted appendage regeneration and wound healing. This indicates that local application could be an optional method as a less invasive prison cell therapy in future. From the observations that ASCs injection stimulated neoangiogenesis at the site of injury, nosotros hypothesize that the grafted ASCs accumulate at the target site and subsequently either differentiate into endothelial cells or raise angiogenesis through paracrine event, leading to wound healing. It has been shown that ASCs can differentiate into endothelial jail cell and better angiogenesis in critical limb ischemia model.26 Further study might be helpful to elucidate the observed ASCs-mediated angiogenesis.

Three weeks after ASCs injection, a significant increment in the number of hair follicles was seen. Although there are reports indicating potential differentiation of stem cells into appendage progenitor cells during skin regeneration, the regenerative outcome of ASCs observed in this study was more probable due to their paracrine signaling.27 , 28 The results of PCNA staining experiments indirectly ostend that ASCs may human activity past accelerating proliferation and differentiation of appendage progenitor cells. ASCs are known to secrete angiogenic growth factors such as HGF and VEGF, which in plow contribute to tissue repair through angiogenetic and antiapoptotic activities.29 , 30 When preincubated with bFGF in vitro, ASCs secrete other growth factors in abundance.thirty Taken together with the results from PCNA staining experiments, these reports led u.s.a. to hypothesize that growth factors secreted from transplanted ASCs is responsible for the skin appendage regeneration seen here.

The mechanism of ASCs in promoting tissue repair is still insufficiently investigated. Information technology is known that ASCs can modulate adverse inflammatory reactions to give positive outcomes. For example, ASCs facilitate tissue regeneration by suppression of TNF-α, IL-6R, and IL-12b.31 Merely TNF-α plays a office on ASCs proliferation, increased expression of proangiogenic factors (FGF-2, VEGF, IL-8, and MCP-1) and inflammatory cytokines (IL-1β, IL-6), and activation of angiogenic and regenerative potential of ASCs.32 In tissue injuries occurring nether astute hypoxic stress conditions, ASCs could upregulate HIF-i, stimulate vascular endothelial growth factor and resistance to significant oxygen deprivation.33

Depending on the beefcake and the extent of damage in the given tissue and organ, the unremarkably used ASCs application routes are either local injections or systemic administration. Bear witness is building for the role of paracrine effects of ASCs in its therapeutic relief, rather than for its effectiveness through local implantation and direct differentiation.34 , 35 Our previous studies have shown that ASCs too exhibit antioxidative and antiapoptotic effects in rescuing tissue from astute ischemic injury.15 In add-on, ASCs may secrete soluble factors such as CINC-1, which activate ERK1/2 and Akt phosphorylation, leading to improved cell survival, tissue repair, and appendage recovery.9 Thus, local injection of ASCs has potential in skin appendage recovery and wound repair.

Delivery of ASCs via diverse routes is dependent on the "native" homing of ASCs to the site of disease or injury.34 , 36 Venous injections of ASCs have been used in treating bladder dysfunction, erectile dysfunction, chronic kidney affliction, and then on.34 , 37 , 38 Intraarterial injection has likewise proved to exist an constructive road of systemic administration. ASCs can be implanted through renal arteries to suppress acute rejection afterward kidney transplantation.39 Cerebral artery apoplexy could also be ameliorated by injection of ASCs via carotid artery.36 Yet, regardless of administration route, the therapeutic efficacy depends on ensuring that sufficient ASCs reside in the damaged area.

Hither, 5 × 105 ASCs transplanted into the subcutaneous space survived in the injection sites throughout the follow-upward period, with a boring transition to deeper subcutaneous adipose tissue layers.twoscore There have been conflicting reports on the number of ASCs used and the results obtained with regards to local ASCs injection. Bliley et al found old application of six.8 × 106 ASCs to be constructive in subeschar injection; increased vascularity, collagen degradation, and dermal adipogenesis were noted following ASCs handling.three In contrast, Loder et al showed that local injection of 1 × 10six ASCs improved burn down wound healing in terms of wound area, wound depth, and apoptotic activity in a mouse model.41 Chang et al demonstrated the therapeutic efficacy of autologous ASCs in treating acute fire wounds in rats by local injection of five × ten6 ASCs.42 Further, Kuo et al injected 1 × 107 ASCs subcutaneously in full-thickness wound of diabetic rats and institute significant subtract in wound healing time.43 Furthermore, Hanson et al showed that i × 106 ASCs, intradermally injected, could ameliorate partial-thickness cutaneous wound healing in porcine model.44 However, another study by Karimi et al reported no meaning improvement utilizing one × 10vi ASCs treatment in acute burn down wound healing (p > 0.05).45 Poor results in the higher up reports may be due to disruption of delicate microenvironment and inflammation and multifocality of many disorders.46 Therefore, optimizing cell delivery is a critical component of a successful cell-based therapy. Hydrogel and collagen scaffold commitment of stem cells have been shown to significantly enhance wound healing.47–50 Further investigations into various routes of cell-commitment including cell-seeded peel substitution needs to be conducted with a view to improve handling outcomes.

In that location are several limitations in the present report. Except road of prison cell delivery and cell engraftment rate, some other limitation is the size and depth of the burn down wounds. Since the allowed environs may substantially differ betwixt major and modest burn wounds, future studies of larger and deeper burn wounds that are better representations of the typical clinical scenario are needed to further evaluate the therapeutic efficacy of ASCs in clinical practice. Our experiments indicated that ASCs could ameliorate the wound healing procedure past angiogenesis and skin appendage regeneration. Nonetheless, the detailed mechanism of ASCs is beyond the scope of this study and to be elucidated. In particular, it is unclear whether these ASCs differentiate into endothelial cells, for example, and become office of the vasculature, or if they play a unlike supportive function at the wound site in promoting neovascularization and wound healing. Currently, our laboratory is conducting further work that addresses this question.

In conclusion, using the new skin isle burn wound rat model, our written report demonstrates that local injection of ASCs could have beneficial therapeutic effects in treating burn wounds. Local injection of ASCs not only improved burn wound recovery merely also enhanced pare appendage regeneration. The healing mechanism may involve ASCs-mediated paracrine stimulation of neoangiogenesis and inhibition of apoptosis.

ACKNOWLEDGMENTS

This written report was supported past Medical Scholarship Foundation in Memory of Professor Albert Ly-Immature Shen.

REFERENCES

ane. Butler KL, Goverman J, Ma H, Fischman A, Yu YM, Bilodeau M, et al Stem cells and burns: review and therapeutic implications. J Fire Care Res. 2010;31:874–81

2. Shin South, Kim Y, Jeong Southward, Hong S, Kim I, Lee W, et al The therapeutic upshot of human adult stem cells derived from adipose tissue in endotoxemic rat model. Int J Med Sci. 2013;x:8–18

3. Bliley JM, Argenta A, Satish 50, McLaughlin MM, Dees A, Tompkins-Rhoades C, et al Assistants of adipose-derived stalk cells enhances vascularity, induces collagen degradation, and dermal adipogenesis in burn wounds. Burns. 2016;42:1212–22

4. Finnerty CC, Przkora R, Herndon DN, Jeschke MG. Cytokine expression contour over time in burned mice. Cytokine. 2009;45:20–v

5. Kim HS, Kim JH, Yim H, Kim D. Changes in the levels of interleukins 6, 8, and 10, tumor necrosis factor alpha, and granulocyte-colony stimulating factor in korean burn down patients: relation to burn size and postburn time. Ann Lab Med. 2012;32:339–44

  • PubMed

vi. Jeschke MG, Gauglitz GG, Finnerty CC, Kraft R, Mlcak RP, Herndon DN. Survivors versus nonsurvivors postburn: differences in inflammatory and hypermetabolic trajectories. Ann Surg. 2014;259:814–23

7. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22

8. Xu J, Woods CR, Mora AL, Joodi R, Brigham KL, Iyer Due south, et al Prevention of endotoxin-induced systemic response by bone marrow-derived mesenchymal stem cells in mice. Am J Physiol Lung Cell Mol Physiol. 2007;293:L131–41

9. Li H, Fu 10, Ouyang Y, Cai C, Wang J, Lord's day T. Adult os-marrow-derived mesenchymal stalk cells contribute to wound healing of peel appendages. Cell Tissue Res. 2006;326:725–36

ten. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, et alTherapeutic Angiogenesis using Cell Transplantation (TACT) Study Investigators. Therapeutic angiogenesis for patients with limb ischaemia past autologous transplantation of bone-marrow cells: a airplane pilot study and a randomised controlled trial. Lancet. 2002;360:427–35

11. Kocher AA, Schuster Md, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, et al Neovascularization of ischemic myocardium by homo bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 2001;7:430–6

12. Rafii Due south, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med. 2003;ix:702–12

13. Moon MH, Kim SY, Kim YJ, Kim SJ, Lee JB, Bae YC, et al Human adipose tissue-derived mesenchymal stem cells ameliorate postnatal neovascularization in a mouse model of hindlimb ischemia. Prison cell Physiol Biochem. 2006;17:279–90

14. Freitag J, Ford J, Bates D, Boyd R, Hahne A, Wang Y, et al Adipose derived mesenchymal stem prison cell therapy in the handling of isolated knee chondral lesions: design of a randomised controlled airplane pilot study comparing arthroscopic microfracture versus arthroscopic microfracture combined with postoperative mesenchymal stem prison cell injections. BMJ Open. 2015;5:e009332

fifteen. Shih YC, Lee PY, Cheng H, Tsai CH, Ma H, Tarng DC. Adipose-derived stem cells exhibit antioxidative and antiapoptotic backdrop to rescue ischemic astute kidney injury in rats. Plast Reconstr Surg. 2013;132:940e–51e

xvi. Chen G, Jin Y, Shi X, Qiu Y, Zhang Y, Cheng M, et al Adipose-derived stem jail cell-based treatment for acute liver failure. Stem Cell Res Ther. 2015;6:40

17. Kim JH, Hong SJ, Park CY, Park JH, Choi SC, Woo SK, et al Intramyocardial adipose-derived stem jail cell transplantation increases pericardial fat with recovery of myocardial role after astute myocardial infarction. Plos One. 2016;11:e0158067

18. Yoon E, Dhar Due south, Chun DE, Gharibjanian NA, Evans GR. In vivo osteogenic potential of man adipose-derived stem cells/poly lactide-co-glycolic acid constructs for bone regeneration in a rat critical-sized calvarial defect model. Tissue Eng. 2007;thirteen:619–27

xix. Lin CH, Yang IC, Tsai CH, Fang HW, Ma H. Auricular tissue technology using osteogenic differentiation of adipose stalk cells with pocket-size intestine submucosa. Plast Reconstr Surg. 2017;140:297–305

twenty. Tobita One thousand, Uysal Air-conditioning, Ogawa R, Hyakusoku H, Mizuno H. Periodontal tissue regeneration with adipose-derived stalk cells. Tissue Eng Part A. 2008;xiv:945–53

  • PubMed

21. Uysal CA, Tobita 1000, Hyakusoku H, Mizuno H. Adipose-derived stem cells enhance primary tendon repair: biomechanical and immunohistochemical evaluation. J Plast Reconstr Aesthet Surg. 2012;65:1712–9

22. Condé-Green A, Marano AA, Lee ES, Reisler T, Cost LA, Milner SM, et al Fat grafting and adipose-derived regenerative cells in burn wound healing and scarring: a systematic review of the literature. Plast Reconstr Surg. 2016;137:302–12

23. Galiano RD, Michaels J fifth, Dobryansky One thousand, Levine JP, Gurtner GC. Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen. 2004;12:485–92

24. Michaels J 5th, Churgin SS, Blechman KM, Greives MR, Aarabi Southward, Galiano RD, et al Db/db mice exhibit severe wound-healing impairments compared with other murine diabetic strains in a silicone-splinted excisional wound model. Wound Repair Regen. 2007;15:665–lxx

25. Davidson JM, Yu F, Opalenik SR. Splinting strategies to overcome confounding wound contraction in experimental animal models. Adv Wound Care (New Rochelle). 2013;ii:142–viii

  • PubMed

26. Miranville A, Heeschen C, Sengenès C, Curat CA, Busse R, Bouloumié A. Comeback of postnatal neovascularization by man adipose tissue-derived stem cells. Circulation. 2004;110:349–55

27. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007;25:2648–59

28. Zhang C, Chen Y, Fu Ten. Sweat gland regeneration later on fire injury: is stem cell therapy a new hope? Cytotherapy. 2015;17:526–35

29. Rehman J, Traktuev D, Li J, Merfeld-Clauss South, Temm-Grove CJ, Bovenkerk JE, et al Secretion of angiogenic and antiapoptotic factors past human adipose stromal cells. Circulation. 2004;109:1292–8

30. Kilroy GE, Foster SJ, Wu X, Ruiz J, Sherwood Due south, Heifetz A, et al Cytokine profile of human adipose-derived stalk cells: expression of angiogenic, hematopoietic, and pro-inflammatory factors. J Cell Physiol. 2007;212:702–9

31. Chang KC, Ma H, Liao WC, Lee CK, Lin CY, Chen CC. The optimal time for early fire wound excision to reduce pro-inflammatory cytokine production in a murine burn injury model. Burns. 2010;36:1059–66

32. Zubkova ES, Beloglazova IB, Makarevich PI, Boldyreva MA, Sukhareva OY, Shestakova MV, et al Regulation of adipose tissue stalk cells angiogenic potential by tumor necrosis factor-blastoff. J Cell Biochem. 2016;117:180–96

33. Andreeva ER, Lobanova MV, Udartseva OO, Buravkova LB. Response of adipose tissue-derived stromal cells in tissue-related O2 microenvironment to short-term hypoxic stress. Cells Tissues Organs. 2015;200:307–fifteen

34. Zhang H, Qiu X, Shindel AW, Ning H, Ferretti L, Jin Ten, et al Adipose tissue-derived stalk cells meliorate diabetic bladder dysfunction in a type II diabetic rat model. Stalk Cells Dev. 2012;21:1391–400

35. Zhang H, Ning H, Banie 50, Wang One thousand, Lin G, Lue TF, et al Adipose tissue-derived stem cells secrete CXCL5 cytokine with chemoattractant and angiogenic properties. Biochem Biophys Res Commun. 2010;402:560–iv

36. Du M, Liu Y, Dang G, Zhu One thousand, Su R, Fan Y, et al Comparison of administration routes for adipose-derived stem cells in the handling of middle cerebral avenue apoplexy in rats. Acta Histochem. 2014;116:1075–84

37. Qiu 10, Villalta J, Ferretti Fifty, Fandel TM, Albersen Chiliad, Lin One thousand, et al Effects of intravenous injection of adipose-derived stem cells in a rat model of radiation therapy-induced erectile dysfunction. J Sex Med. 2012;9:1834–41

38. Quimby JM, Webb TL, Randall E, Marolf A, Valdes-Martinez A, Dow SW. Assessment of intravenous adipose-derived allogeneic mesenchymal stalk cells for the treatment of feline chronic kidney disease: a randomized, placebo-controlled clinical trial in eight cats. J Feline Med Surg. 2016;18:165–71

39. Kato T, Okumi M, Tanemura K, Yazawa K, Kakuta Y, Yamanaka Grand, et al Adipose tissue-derived stalk cells suppress acute cellular rejection by TSG-6 and CD44 interaction in rat kidney transplantation. Transplantation. 2014;98:277–84

40. Koellensperger E, Lampe K, Beierfuss A, Gramley F, Germann G, Leimer U. Intracutaneously injected human adipose tissue-derived stem cells in a mouse model stay at the site of injection. J Plast Reconstr Aesthet Surg. 2014;67:844–50

41. Loder S, Peterson JR, Agarwal Southward, Eboda O, Brownley C, DeLaRosa S, et al Wound healing after thermal injury is improved by fat and adipose-derived stem jail cell isografts. J Burn Intendance Res. 2015;36:70–6

42. Chang YW, Wu YC, Huang SH, Wang HD, Kuo Twelvemonth, Lee SS. Autologous and not allogeneic adipose-derived stem cells improve acute burn wound healing. Plos One. 2018;13:e0197744

43. Kuo Twelvemonth, Wang CT, Cheng JT, Kao GS, Chiang YC, Wang CJ. Adipose-derived stalk cells accelerate diabetic wound healing through the induction of autocrine and paracrine furnishings. Prison cell Transplant. 2016;25:71–81

44. Hanson SE, Kleinbeck KR, Cantu D, Kim J, Bentz ML, Faucher LD, et al Local commitment of allogeneic bone marrow and adipose tissue-derived mesenchymal stromal cells for cutaneous wound healing in a porcine model. J Tissue Eng Regen Med. 2016;10:E90–100

  • PubMed

45. Karimi H, Soudmand A, Orouji Z, Taghiabadi Eastward, Mousavi SJ. Burn wound healing with injection of adipose-derived stem cells: a mouse model written report. Ann Burns Fire Disasters. 2014;27:44–nine

  • PubMed

46. Khaldoyanidi S. Directing stem cell homing. Prison cell Stem Cell. 2008;2:198–200

  • PubMed

47. Natesan Due south, Zamora DO, Wrice NL, Baer DG, Christy RJ. Bilayer hydrogel with autologous stem cells derived from debrided man fire skin for improved skin regeneration. J Burn Care Res. 2013;34:18–30

48. Foubert P, Barillas S, Gonzalez Advert, Alfonso Z, Zhao Southward, Hakim I, et al Uncultured adipose-derived regenerative cells (adrcs) seeded in collagen scaffold improves dermal regeneration, enhancing early vascularization and structural arrangement following thermal burns. Burns. 2015;41:1504–16

49. Chung E, Rybalko VY, Hsieh PL, Leal SL, Samano MA, Willauer AN, et al Fibrin-based stem cell containing scaffold improves the dynamics of fire wound healing. Wound Repair Regen. 2016;24:810–9

50. Burmeister DM, Stone R 2nd, Wrice N, Laborde A, Becerra SC, Natesan S, et al Delivery of allogeneic adipose stem cells in polyethylene glycol-fibrin hydrogels as an adjunct to meshed autografts later abrupt debridement of deep partial thickness burns. Stem Cells Transl Med. 2018;7:360–72

Keywords:

Adipose-derived stalk cell; Angiogenesis; Burn down; Rat; Peel appendage; Wound healing

© 2019 by Lippincott Williams & Wilkins, Inc.

Source: https://journals.lww.com/jcma/fulltext/2019/08000/adipose_derived_stem_cells_induced_burn_wound.8.aspx

Posted by: floresanion1954.blogspot.com

0 Response to "Is It Necessary To Grow Human Skin On A Rat For Burn Victims?"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel