Acessibilidade / Reportar erro

Stem cells in dermatology* * Work performed at Hemocord - Stem Cell Bank - Porto Alegre, RS, Brazil.

Abstract

Preclinical and clinical research have shown that stem cell therapy could be a promising therapeutic option for many diseases in which current medical treatments do not achieve satisfying results or cure. This article describes stem cells sources and their therapeutic applications in dermatology today.

Adult stem cells; Dermatology; Hematopoietic stem cells; Stem cells; Regenerative medicine


INTRODUCTION

The current trend in medicine is to focus on two major areas in which until recently there was not much emphasis on: "prevention" of diseases and "regenerative medicine". The first provides the power to change an individual's destiny, preventing a disease from occurring and increasing life expectancy, while the second is an attempt to cure diseases for which modern medicine has yet no treatment available. This article aims to briefly address regenerative medicine, with the theme "stem cells", from its discovery to its current applications and future prospects, describing the important role of skin cells in this context.

ORIGIN AND CONCEPTS

Bone marrow transplantation for hematologic diseases occurs since the 1950s.11. Thomas ED, Lochte HL, Lu WC, Ferrebee JW. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N Engl J Med. 1957;257:491-6. It was known then, that there was a type of cell within the bone marrow that could give rise to all lineages of blood cells, determining the success of this treatment.11. Thomas ED, Lochte HL, Lu WC, Ferrebee JW. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N Engl J Med. 1957;257:491-6. Thus emerged the first concept of a primitive cell that specialized to generate other cells with specific functions. At the end of the 90s, came the first evidence that, faced with different stimuli and environments, this hematopoietic progenitor cell could generate cells that differed from the original tissue (plasticity) and that they were attracted to damaged tissues distant from their surroundings.22. Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, Stornaiuolo A, Cossu G, et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science. 1998;279:1528-30.,33. Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, Murase N, et al. Bone marrow as a potential source of hepatic oval cells. Science. 1999;284:1168-70.,44. Eglitis MA, Mezey E. Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc Natl Acad Sci U S A. 1997;94:4080-5.,55. Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238:265-72. Cells that presented this behavior were named stem cells.

For a cell to be considered a stem cell, it has to present three characteristics: self-renewal, i.e., asymmetric division resulting both in cells that are similar to it, as well as specialized cells; the ability to regenerate the tissue in which it is located, and plasticity, that is the ability to generate other cell types, different from those of the original tissue.66. Herzog EL, Chai L, Krause DS. Plasticity of marrow-derived stem cells. Blood. 2003;102:3483-93.

From the moment that stem cells were discovered in bone marrow, the search for other sources began. Fundamentally, stem cells are present in various body tissues, from the embryo to the adult individual, since all tissues have some degree of repair capacity.77. Pretheeban T, Lemos DR, Paylor B, Zhang RH, Rossi FM. Role of stem/progenitor cells in reparative disorders. Fibrogenesis Tissue Repair. 2012;5:20. One of the most comprehensive classifications divides them in two groups: embryonic stem cells present in the blastocyst, the inner cell mass of an embryo five days after fertilization of the egg by the sperm; and adult stem cells present from the formation of the fetus and responsible for repairing injured tissues composed of cells in more specialized stages (Figure 1).88. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145-7.,99. Trounson A. Human embryonic stem cells: mother of all cell and tissue types. Reprod Biomed Online. 2002;4:58-63.,1010. Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006;311:1880-5.,1111. Broxmeyer HE, Douglas GW, Hangoc G, Cooper S, Bard J, English D, et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci U S A. 1989;86:3828-32. Amongst adult stem cells there are two types that differ in their origin and differentiation capacity: the aforementioned hematopoietic stem cells, originally derived from bone marrow that give rise to all hematopoietic tissues, and mesenchymal stem cells pre-determined to differentiate into various cell lineages of mesodermal origin such as osteogenic, chondrogenic and adipogenic lineages, and cartilage and bone tissues.1010. Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006;311:1880-5.,1111. Broxmeyer HE, Douglas GW, Hangoc G, Cooper S, Bard J, English D, et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci U S A. 1989;86:3828-32.,1212. Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise review: role of mesenchymal stem cells in wound repair. Stem Cells Transl Med. 2012;1:142-9. These cells have a significant role in tissue repair, homeostasis and immunomodulation. Some tissues are rich in mesenchymal stem cells and are easily harvested, such as the adipose tissue, dermis, bone marrow and umbilical cord tissue, including Wharton's jelly.1313. Batsali AK, Kastrinaki MC, Papadaki HA, Pontikoglou C. Mesenchymal Stem Cells derived from Wharton's Jelly of the Umbilical Cord: biological properties and emerging clinical applications. Curr Stem Cell Res Ther. 2013;8:144-55.,1414. Karagianni M, Brinkmann I, Kinzebach S, Grassl M, Weiss C, Bugert P, et al. A comparative analysis of the adipogenic potential in human mesenchymal stromal cells from cord blood and other sources. Cytotherapy. 2013;15:76-88. Both types of adult stem cells, despite having a pre-determined differentiation into certain tissues, demonstrated in vitro ability to transform, among others, into neuronal, hepatic, and muscular tissues. Mesenchymal stem cells are included in more than 200 clinical trials for various diseases like diabetes, ulcerative colitis, systemic lupus erythematosus, dilated cardiomyopathy, cirrhosis, spinal cord injury, and osteoarthritis to cite a few. Hematopoietic stem cells, from bone marrow and umbilical cord blood, are being evaluated for hematologic and non-hematologic diseases in over 3,000 clinical studies with protocols registered in the United States National Institute of Health Program ClinicalTrials.gov (www.clinicaltrials.gov).

FIGURE 1:
Origin of adult human tissues from embryonic stem cells. Differentiation and maturation of cells can occur in the embryonic and adult periods. Embryonic stem cells can originate tissues from all three germ layers. The mesodermal layer produces mesenchymal and hematopoietic precursor cells. Adult tissues have natural stocks of resident stem cells, and in the cellular differentiation cascade there are cells going to undescribed differentiation processes. The main sources of embryonic and adult stem cells for therapeutic purposes are presented below

MAIN SOURCES OF ADULT STEM CELLS

Organs that have a significant degree of cell turnover, such as bone marrow and skin, have a tendency to present cell populations richer in stem cells.

Alternatives for easy retrieval and storage of hematopoietic stem cells in high concentration are the bone marrow and umbilical cord and placental blood, the latter being the only form to collect in which there is no need for surgical intervention, since it is drawn after clamping the umbilical cord concomitanlty or after the manual removal of the placenta.1111. Broxmeyer HE, Douglas GW, Hangoc G, Cooper S, Bard J, English D, et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci U S A. 1989;86:3828-32. There are several advantages described for stem cells derived from cord blood, such as lower viral disease transmission's incidence, high regenerative power and low immunogenicity, all secondary to the time of birth when there is less exposure to external agents and the fact that the newborn is immunologically immature.1515. Gluckman E, Rocha V, Boyer-Chammard A, Locatelli F, Arcese W, Pasquini R, et al. Outcome of cord-blood transplantation from related and unrelated donors. Eurocord Transplant Group and the European Blood and Marrow Transplantation Group. N Engl J Med. 1997;337:373-81. Bone marrow, adipose tissue, dermis and umbilical cord tissue are all sources with high concentration of mesenchymal stem cells.1616. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211-28. Among these, we highlight the umbilical cord tissue, which can be collected in a non-invasive manner, and has cellular regenerative potential comparable to the regenerative power of the skin of a very young individual.1313. Batsali AK, Kastrinaki MC, Papadaki HA, Pontikoglou C. Mesenchymal Stem Cells derived from Wharton's Jelly of the Umbilical Cord: biological properties and emerging clinical applications. Curr Stem Cell Res Ther. 2013;8:144-55.

Until the present, only hematopoietic stem cells from bone marrow and umbilical cord blood are authorized for medical use. They may be used in hematological, genetic or acquired diseases, and also some common childhood tumors such as neuroblastoma, retinoblastoma, Wilms' tumor, and osteosarcoma.1717. Barriga F, Ramírez P, Wietstruck A, Rojas N. Hematopoietic stem cell transplantation: clinical use and perspectives. Biol Res. 2012;45:307-16.

STEM CELLS AND SKIN

The skin, being an organ of great cell replication, has several groups of stem cells present in its layers.

Interfollicular stem cells are found in the epidermis, near the basal membrane. Their primary role is to repair epidermal trauma. In the hair follicle's histologic complex there are follicular, sebaceous and neural crest stem cells. Follicular and neural crest stem cells cohabit in the bulge. In the sebaceous gland, we can find sebaceous stem cells.1818. Blanpain C, Fuchs E. Epidermal stem cells of the skin. Annu Rev Cell Dev Biol. 2006;22:339-73.,1919. Fuchs E. Skin stem cells: rising to the surface. J Cell Biol. 2008;180:273-84.

Stem cells within the dermis, adipose tissue and hypodermis are essentially of mesodermal origin, therefore mesenchymal. They are closely associated with blood vessels and generate fibroblasts and myofibroblasts that participate actively in repair mechanisms. Mesenchymal stem cells have a strong link with repair and regeneration processes in soft tissue, musculoskeletal and vascular systems.2020. Hill RP, Gledhill K, Gardner A, Higgins CA, Crawford H, Lawrence C, et al. Generation and characterization of multipotent stem cells from established dermal cultures. PLoS One. 2012;7:e50742.,2121. Chang P, Tao K. The use of adipose tissue-derived stem cells within a dermal substitute improves skin regeneration by increasing neoangiogenesis and collagen synthesis. Plast Reconstr Surg. 2013;131:116e-7e.

There is still no definite pattern, which can prioritize and define exactly what degree of importance a cell has, compared to another, in the skin repair mechanism. However it is known that, there is a marked interplay between systems through molecular interactions, such as the cells of the hypodermis presenting paracrine action over dermal fibroblasts. Mesenchymal stem cells located in the dermis and hypodermis are critical in this process because they coordinate the response of tissue repair by recruiting other host cells, growth factors and extracellular matrix secretory proteins.1212. Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise review: role of mesenchymal stem cells in wound repair. Stem Cells Transl Med. 2012;1:142-9.,2222. Alfaro MP, Deskins DL, Wallus M, DasGupta J, Davidson JM, Nanney LB, et al. A physiological role for connective tissue growth factor in early wound healing. Lab Invest. 2013;93:81-95.

ROLE OF CUTANEOUS STEM CELLS IN THE REGENERATION OF OTHER TISSUES

Cutaneous stem cells have been experimentally explored, in several animal models and in vitro, to demonstrate their potential and plasticity, especially because the skin is an inexhaustible source of stem cells that are easy to obtain; besides being autologous cells, thereby avoiding complications such as graftversus-host disease.

Due to the ontologic proximity between neural tube stem cells and follicular complex's neural crest cells, they may be used in cell therapy for spinal cord injuries. Sieber-Blum from the University of Newcastle, in a trial with mice, observed an improvement of 24% in sensitivity and perception of touch in the lower limbs after induced spinal cord injuries.2323. Sieber-Blum M Epidermal neural crest stem cells and their use in mouse models of spinal cord injury. Brain Res Bull. 2010;83:189-93. Although cell transplantation was performed unilaterally, improvements were bilateral, suggesting that genes from epidermal neural crest cells encode and express neutrophins, and trophic and angiogenic factors, that justify bilateral functional improvements. Recently, researchers from the same center demonstrated the multipotentiality of human epidermal cells form the neural crest, through the isolation, characterization and ex-vivo expansion, transforming them into osteocytes and melanocytes, thus identifying a source of easy access and great power of differentiation.2424. Sieber-Blum M, Hu Y. Epidermal neural crest stem cells (EPI-NCSC) and pluripotency. Stem Cell Rev. 2008;4:256-60.,2525. Clewes O, Narytnyk A, Gillinder KR, Loughney AD, Murdoch AP, Sieber-Blum M. Human epidermal neural crest stem cell characterization and direct differentiation into osteocytes and melanocytes. Stem Cell Rev. 2011;7:799-814.

Mesenchymal stem cells from human dermis have also shown great power of expansion in vitro, especially when collected from newborns. Their easy obtainment, multiplication and security, seen in population doublings in culture, strongly suggests that in the future they may be conveniently used for the regeneration of tissues, such as adipose, muscle and osteogenic.2626. Bartsch G, Yoo JJ, De Coppi P, Siddiqui MM, Schuch G, Pohl HG, et al. Propagation, Expansion, and Multilineage Differentiation of Human Somatic Stem Cells from Dermal Progenitors - muscle, adipose tissue and bone Stem Cells Dev. 2005;14:337-48.

Since 2006, dermal fibroblasts have been manipulated in vitro and genetically reprogrammed to regress to an immature and undifferentiated state that precedes their current state of differentiation; afterwards they were induced to develop into various cell lines. These immature cells produced in vitro, derived from fibroblast regression, are called induced pluripotent cells (iPS).2727. Teoh HK, Cheong SK. Induced pluripotent stem cells in research and therapy. Malays J Pathol. 2012;34:1-13.

Surprisingly, dermal fibroblasts themselves have also demonstrated characteristics of in vitro pluripotency, without the need to be induced to immaturity by the activation of embryonic stage genes. Canadian researchers obtained a hematopoietic progenitor cell from a fibroblast through the application of specific cytokines. This hematopoietic precursor cell, developed in vitro, was able to generate granulocytic, monocytic, megakaryocytic and erythroid lineages, besides demonstrating the ability to repopulate the bone marrow by grafting.2828. Szabo E, Rampalli S, Risueño RM, Schnerch A, Mitchell R, Fiebig-Comyn A, et al. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature. 2010;468:521-6.

INDUCED PLURIPOTENT CELLS (iPS)

As described earlier, skin cells have a significant role in stem cell studies, particullarly in the retrieval of induced pluripotent cells, which are an alternative to using embryonic stem cells or therapeutic cloning in research.

In 2006, Japanese researchers genetically reprogrammed cells from mouse tail, so that they reverted to the behaviour of embryonic stem cells. This reprogramming process occurs through the insertion of a virus containing four genes. These genes are inserted into the DNA of an adult cell (e.g. skin cell) and reprogram its genetic code. With this new program, the cells return to the stage of an embryonic stem cell, with characteristics of self-renewal and the ability to differentiate into any tissue.2929. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-76.

Later, in 2007, the first human induced cells developed from skin cells were produced. This has been so far the main source of cells for reprogramming.3030. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861-72.,3131. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917-20. This type of cell lineage brings benefits such as the ability to generate cells with different disease models, in order to study pathophysiological mechanisms and test new drugs, and without ethical conflicts such as those that occur with the use of embryonic stem cells. Another benefit is that they can be generated from the patient and possibly be used as a source for autologous cell therapy. This eliminates the risk of rejection increasing the chance of successful transplantation.3232. Longstaff H, McDonald M, Bailey J. Communicating Risks and Benefits About Ethically Controversial Topics: the Case of Induced Pluripotent Stem (iPS) Cells. Stem Cell Rev. 2013;9:388-96.

Numerous healthy cell lines have been developed from induced pluripotent stem cells, such as cardiomyocytes and liver cells, and for many disease models as well, like Alzheimer's, type 1 diabetes, cerebellar ataxia, and others. Since iPSs production technique has currently been mastered, studies now focus on the safety of their use in clinical therapies.3333. Xia G, Santostefano K, Hamazaki T, Liu J, Subramony SH, Terada N, et al. Generation of Human-Induced Pluripotent Stem Cells to Model Spinocerebellar Ataxia Type 2 In vitro. J Mol Neurosci. 2013;51:237-48.

34. Iwamuro M, Shiraha H, Nakaji S, Furutani M, Kobayashi N, Takaki A, et al. A preliminary study for constructing a bioartificial liver device with induced pluripotent stem cell-derived hepatocytes. Biomed Eng Online. 2012;11:93.

35. Jiang B, Dong H, Li Q, Yu Y, Zhang Z, Zhang Y, et al. Differentiation of Reprogrammed Mouse Cardiac Fibroblasts into Functional Cardiomyocytes. Cell Biochem Biophys. 2013;66:309-18.

36. Yahata N, Asai M, Kitaoka S, Takahashi K, Asaka I, Hioki H, et al. Anti-Aß drug screening platform using human iPS cell-derived neurons for the treatment of Alzheimer's disease. PLoS One. 2011;6:e25788.
-3737. Thatava T, Kudva YC, Edukulla R, Squillace K, De Lamo JG, Khan YK, et al. Intrapatient Variations in Type 1 Diabetes-specific iPS Cell Differentiation Into Insulin-producing Cells. Mol Ther. 2013;21:228-39.

OTHER ADULT STEM CELL SOURCES FOR CUTANEOUS DISEASES

When it comes to treatment of cutaneous diseases, the use of cellular therapy with stem cells, regardless of the source of cells used, is still considered experimental.

Nonetheless, several clinical protocols are in progress, such as allogeneic bone marrow transplantation for recessive dystrophic epidermolysis bullosa. After results of preclinical studies showing significant improvement in the presence of collagen VII in mice, a clinical study of allogeneic stem cell transplantation demonstrated increased production of collagen VII in the host and presence of donor cells in the recipient's skin.3838. Wagner JE, Ishida-Yamamoto A, McGrath JA, Hordinsky M, Keene DR, Woodley DT, et al. Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. (fase 1). N Engl J Med. 2010;363:629-39 More studies are needed to monitor disease progression and assess risks and benefits of this therapy.

Comparing the hematopoietic stem cells nonablative autologous transplantation versus the traditional treatment with cyclophosphamide for systemic sclerosis, a randomized study demonstrated improvement of skin and also pulmonary function for at least two years post transplantation, when compared with conventional treatment.3939. Burt RK, Shah SJ, Dill K, Grant T, Gheorghiade M, Schroeder J, et al. Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): an openlabel, randomised phase 2 trial. Lancet. 2011;378:498-506.

Autologous hematopoietic progenitor cells transplantation has been considered an experimental therapeutic alternative for systemic lupus erythematosus with systemic symptoms and those still refractory to conventional treatments. Transplantation using mesenchymal stem cells from bone marrow in autoimmune diseases like lupus have resulted in clinical remission and functional improvement of the affected organs.4040. Wang D, Zhang H, Liang J, Li X, Feng X, Wang H, et al. Allogeneic mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus: 4 years experience. Cell Transplant. 2012 Oct 31. . [Epub ahead of print] More studies are needed for the use of these alternative therapies on a larger scale.

STEM CELLS IN CHRONIC WOUNDS

Chronic wounds can be considered an ever growing problem, since nowadays 50% of severe skin wounds do not respond to current treatments.4141. Mustoe TA, O'Shaughnessy K, Kloeters O. Chronic wound pathogenesis and current treatment strategies: A unifying hypothesis. Plast Reconstr Surg. 2006;117:35S-41S. Wound healing is a complex process that requires a composition of factors equivalent that of native skin and also a coordinated interplay of extra-cellular matrix, growth factors, cells and endogenous proteins.4242. Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise Review: Role of Mesenchymal Stem Cells in Wound Repair. Stem Cells Transl Med. 2012;1:142-9.

The exogenous application of stem cells for wound healing can be considered a promising solution because of their intrinsic capacity to self-renew and differentiate into various tissues.4343. Chan WK, Lau AS, Li JC, Law HK, Lau YL, Chan GC. MHC expression kinetics and immunogenicity of mesenchymal stromal cells after short-term IFN-gamma challenge. Exp Hematol. 2008;36:1545-55. The use of stem cells in wound healing is clinically relevant, especially in those that are difficult to heal, such as lesions resulting from diabetes, major trauma, vascular insufficiency, severe and extensive burns and numerous other conditions.4242. Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise Review: Role of Mesenchymal Stem Cells in Wound Repair. Stem Cells Transl Med. 2012;1:142-9.

Mesenchymal stem cells play an important role in the remodeling of wound healing, being assigned to them paracrine factors with potential to promote improvement in the general state of a given tissue damage or injury recovery in various degrees. These paracrine effects are described as immunomodulating, anti-apoptotic, pro-angiogenic, chemo-attractor, and anti-fibrosis effects and also the support to endogenous stem cells growth and differentiation.4444. Karlsson H, Samarasinghe S, Ball LM, Sundberg B, Lankester AC, Dazzi F, et al. Mesenchymal stem cells exert differential effects on alloantigen and virus-specific T-cell responses. Blood. 2008;112:532-41.,4545. Meirelles Lda S, Nardi NB. Methodology, biology and clinical applications of mesenchymal stem cells. Front Biosci (Landmark Ed). 2009;14:4281-98. The presence of mesenchymal stem cells at the site of injury directs the regeneration and consequently the return of natural physiological functioning leading to regenerative success.4242. Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise Review: Role of Mesenchymal Stem Cells in Wound Repair. Stem Cells Transl Med. 2012;1:142-9.

Although bone marrow is one of the most frequently used sources for obtaining the stem cells used in cell therapy for wounds regeneration, various sources have been successfully adopted, including skin, adipose tissue, periosteum, tendons, muscle, and others. A source which recently became a target for research is the use of tissue extracted from placenta and human umbilical cord. Comparison of mesenchymal stem cells derived from bone marrow with those derived from placenta or umbilical cord tissue showed minimal differences in cellular phenotype, differentiation and other properties assigned to them.4646. Barlow S, Brooke G, Chatterjee K, Price G, Pelekanos R, Rossetti T, et al. Comparison of human placenta- and bone marrow-derived multipotent mesenchymal stem cells. Stem Cells Dev. 2008;17:1095-107.,4747. Poloni A, Rosini V, Mondini E, Maurizi G, Mancini S, Discepoli G, et al. Characterization and expansion of mesenchymal progenitor cells from first-trimester chorionic villi of human placenta. Cytotherapy. 2008;10:690-7.

Mesenchymal stem cells play an important role in mediating each stage of the wound healing process. During the inflammatory phase, they can coordinate the effects of inflammation by stimulating anti-inflammatory cytokines, as well as inhibiting the deleterious effects of pro-inflammatory cytokines. This ability to promote the attenuation of inflammation is particularly critical for chronic wound treatment, in which high levels of inflammation can prevent the tissue regeneration process. Mesenchymal stem cells contribute to the proliferative phase of regeneration through the secretion of growth factors such as VEGF, bFGF, KGF and promotion of granulation and epithelialization. These cells can also regulate remodeling of the healed wound by promoting organized extracellular matrix deposition during its replacement.4242. Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise Review: Role of Mesenchymal Stem Cells in Wound Repair. Stem Cells Transl Med. 2012;1:142-9.

The advantages of using mesenchymal stem cells for wound healing were demonstrated in various preclinical and clinical studies. Although many products are currently available to treat severe wounds, existing therapies are still inadequate in many cases, and mesenchymal stem cells are an attractive alternative to promote regeneration.

STEM CELLS AND MELANOMA

Knowledge on the pathophysiology of melanoma's origins is changing. Traditionally, it was assumed that cancer cells arose from melanocytes. Due to the presence of melanocyte precursors in the dermis, an hypothesis was formulated that melanoma could also originate in extrafollicular stem cells modified by harming factors such as UVA and UVB.4848. Hoerter JD, Bradley P, Casillas A, Chambers D, Denholm C, Johnson K, et al. Extrafollicular dermal melanocyte stem cells and melanoma. Stem Cells Int. 2012;2012:407079. Therefore, mechanisms that lead to melanoma formation may occur by cellular changes in melanocytes, melanocytes'stem cell precursors or both. Experimental studies are underway to elucidate the mechanisms capable of causing damage to the DNA of stem cells, to ascertain this hypothesis.4949. Hoerter JD, Bradley P, Casillas A, Chambers D, Weiswasser B, Clements L, et al. Does Melanoma Begin in a Melanocyte Stem Cell? J Skin Cancer. 2012;2012:571087.

Currently, clinical trials using hematopoietic stem cells transplantation, adjuvant to chemotherapy and immunotherapy for patients with metastatic melanoma, are underway in several centers, however without definitive results.

Hematopoietic precursor cells transplant would allow the use of higher doses of chemotherapy for superior eradication of tumour cells (www.clinicaltrials.gov).

CONCLUSION

Hematopoietic stem cells have contributed directly to the health of patients currently treating hematological diseases such as leukemia and lymphoma, metabolic and immunologic disorders, besides the most prevalent solid tumors in childhood. However, its potential for the treatment of diseases that depend on significant and specialized tissue regeneration is very great and has been evidenced by the scientific community through the numerous clinical trials underway. Among these trials are researches on cutaneous diseases with systemic repercussions, showing promising results. Stem cells located in the skin have shown interesting plasticity, moreover, mesenchymal cells of the dermis, hypodermis and other sources are involved in studies to promote chronic wounds regeneration. Induced pluripotent stem cells are mostly produced using skin cells and they play an important role in the development of cell lineages that allow the study of certain diseases' pathophysiology and test new drugs.

The scientific community strives to unravel the potential diversity of stem cells present in the tissues and determine the best sources for specific diseases, and certainly skin stem cells have played an important role in this process.

REFERENCES

  • 1
    Thomas ED, Lochte HL, Lu WC, Ferrebee JW. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N Engl J Med. 1957;257:491-6.
  • 2
    Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, Stornaiuolo A, Cossu G, et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science. 1998;279:1528-30.
  • 3
    Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, Murase N, et al. Bone marrow as a potential source of hepatic oval cells. Science. 1999;284:1168-70.
  • 4
    Eglitis MA, Mezey E. Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc Natl Acad Sci U S A. 1997;94:4080-5.
  • 5
    Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238:265-72.
  • 6
    Herzog EL, Chai L, Krause DS. Plasticity of marrow-derived stem cells. Blood. 2003;102:3483-93.
  • 7
    Pretheeban T, Lemos DR, Paylor B, Zhang RH, Rossi FM. Role of stem/progenitor cells in reparative disorders. Fibrogenesis Tissue Repair. 2012;5:20.
  • 8
    Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145-7.
  • 9
    Trounson A. Human embryonic stem cells: mother of all cell and tissue types. Reprod Biomed Online. 2002;4:58-63.
  • 10
    Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006;311:1880-5.
  • 11
    Broxmeyer HE, Douglas GW, Hangoc G, Cooper S, Bard J, English D, et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci U S A. 1989;86:3828-32.
  • 12
    Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise review: role of mesenchymal stem cells in wound repair. Stem Cells Transl Med. 2012;1:142-9.
  • 13
    Batsali AK, Kastrinaki MC, Papadaki HA, Pontikoglou C. Mesenchymal Stem Cells derived from Wharton's Jelly of the Umbilical Cord: biological properties and emerging clinical applications. Curr Stem Cell Res Ther. 2013;8:144-55.
  • 14
    Karagianni M, Brinkmann I, Kinzebach S, Grassl M, Weiss C, Bugert P, et al. A comparative analysis of the adipogenic potential in human mesenchymal stromal cells from cord blood and other sources. Cytotherapy. 2013;15:76-88.
  • 15
    Gluckman E, Rocha V, Boyer-Chammard A, Locatelli F, Arcese W, Pasquini R, et al. Outcome of cord-blood transplantation from related and unrelated donors. Eurocord Transplant Group and the European Blood and Marrow Transplantation Group. N Engl J Med. 1997;337:373-81.
  • 16
    Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211-28.
  • 17
    Barriga F, Ramírez P, Wietstruck A, Rojas N. Hematopoietic stem cell transplantation: clinical use and perspectives. Biol Res. 2012;45:307-16.
  • 18
    Blanpain C, Fuchs E. Epidermal stem cells of the skin. Annu Rev Cell Dev Biol. 2006;22:339-73.
  • 19
    Fuchs E. Skin stem cells: rising to the surface. J Cell Biol. 2008;180:273-84.
  • 20
    Hill RP, Gledhill K, Gardner A, Higgins CA, Crawford H, Lawrence C, et al. Generation and characterization of multipotent stem cells from established dermal cultures. PLoS One. 2012;7:e50742.
  • 21
    Chang P, Tao K. The use of adipose tissue-derived stem cells within a dermal substitute improves skin regeneration by increasing neoangiogenesis and collagen synthesis. Plast Reconstr Surg. 2013;131:116e-7e.
  • 22
    Alfaro MP, Deskins DL, Wallus M, DasGupta J, Davidson JM, Nanney LB, et al. A physiological role for connective tissue growth factor in early wound healing. Lab Invest. 2013;93:81-95.
  • 23
    Sieber-Blum M Epidermal neural crest stem cells and their use in mouse models of spinal cord injury. Brain Res Bull. 2010;83:189-93.
  • 24
    Sieber-Blum M, Hu Y. Epidermal neural crest stem cells (EPI-NCSC) and pluripotency. Stem Cell Rev. 2008;4:256-60.
  • 25
    Clewes O, Narytnyk A, Gillinder KR, Loughney AD, Murdoch AP, Sieber-Blum M. Human epidermal neural crest stem cell characterization and direct differentiation into osteocytes and melanocytes. Stem Cell Rev. 2011;7:799-814.
  • 26
    Bartsch G, Yoo JJ, De Coppi P, Siddiqui MM, Schuch G, Pohl HG, et al. Propagation, Expansion, and Multilineage Differentiation of Human Somatic Stem Cells from Dermal Progenitors - muscle, adipose tissue and bone Stem Cells Dev. 2005;14:337-48.
  • 27
    Teoh HK, Cheong SK. Induced pluripotent stem cells in research and therapy. Malays J Pathol. 2012;34:1-13.
  • 28
    Szabo E, Rampalli S, Risueño RM, Schnerch A, Mitchell R, Fiebig-Comyn A, et al. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature. 2010;468:521-6.
  • 29
    Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-76.
  • 30
    Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861-72.
  • 31
    Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917-20.
  • 32
    Longstaff H, McDonald M, Bailey J. Communicating Risks and Benefits About Ethically Controversial Topics: the Case of Induced Pluripotent Stem (iPS) Cells. Stem Cell Rev. 2013;9:388-96.
  • 33
    Xia G, Santostefano K, Hamazaki T, Liu J, Subramony SH, Terada N, et al. Generation of Human-Induced Pluripotent Stem Cells to Model Spinocerebellar Ataxia Type 2 In vitro. J Mol Neurosci. 2013;51:237-48.
  • 34
    Iwamuro M, Shiraha H, Nakaji S, Furutani M, Kobayashi N, Takaki A, et al. A preliminary study for constructing a bioartificial liver device with induced pluripotent stem cell-derived hepatocytes. Biomed Eng Online. 2012;11:93.
  • 35
    Jiang B, Dong H, Li Q, Yu Y, Zhang Z, Zhang Y, et al. Differentiation of Reprogrammed Mouse Cardiac Fibroblasts into Functional Cardiomyocytes. Cell Biochem Biophys. 2013;66:309-18.
  • 36
    Yahata N, Asai M, Kitaoka S, Takahashi K, Asaka I, Hioki H, et al. Anti-Aß drug screening platform using human iPS cell-derived neurons for the treatment of Alzheimer's disease. PLoS One. 2011;6:e25788.
  • 37
    Thatava T, Kudva YC, Edukulla R, Squillace K, De Lamo JG, Khan YK, et al. Intrapatient Variations in Type 1 Diabetes-specific iPS Cell Differentiation Into Insulin-producing Cells. Mol Ther. 2013;21:228-39.
  • 38
    Wagner JE, Ishida-Yamamoto A, McGrath JA, Hordinsky M, Keene DR, Woodley DT, et al. Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. (fase 1). N Engl J Med. 2010;363:629-39
  • 39
    Burt RK, Shah SJ, Dill K, Grant T, Gheorghiade M, Schroeder J, et al. Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): an openlabel, randomised phase 2 trial. Lancet. 2011;378:498-506.
  • 40
    Wang D, Zhang H, Liang J, Li X, Feng X, Wang H, et al. Allogeneic mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus: 4 years experience. Cell Transplant. 2012 Oct 31. . [Epub ahead of print]
  • 41
    Mustoe TA, O'Shaughnessy K, Kloeters O. Chronic wound pathogenesis and current treatment strategies: A unifying hypothesis. Plast Reconstr Surg. 2006;117:35S-41S.
  • 42
    Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise Review: Role of Mesenchymal Stem Cells in Wound Repair. Stem Cells Transl Med. 2012;1:142-9.
  • 43
    Chan WK, Lau AS, Li JC, Law HK, Lau YL, Chan GC. MHC expression kinetics and immunogenicity of mesenchymal stromal cells after short-term IFN-gamma challenge. Exp Hematol. 2008;36:1545-55.
  • 44
    Karlsson H, Samarasinghe S, Ball LM, Sundberg B, Lankester AC, Dazzi F, et al. Mesenchymal stem cells exert differential effects on alloantigen and virus-specific T-cell responses. Blood. 2008;112:532-41.
  • 45
    Meirelles Lda S, Nardi NB. Methodology, biology and clinical applications of mesenchymal stem cells. Front Biosci (Landmark Ed). 2009;14:4281-98.
  • 46
    Barlow S, Brooke G, Chatterjee K, Price G, Pelekanos R, Rossetti T, et al. Comparison of human placenta- and bone marrow-derived multipotent mesenchymal stem cells. Stem Cells Dev. 2008;17:1095-107.
  • 47
    Poloni A, Rosini V, Mondini E, Maurizi G, Mancini S, Discepoli G, et al. Characterization and expansion of mesenchymal progenitor cells from first-trimester chorionic villi of human placenta. Cytotherapy. 2008;10:690-7.
  • 48
    Hoerter JD, Bradley P, Casillas A, Chambers D, Denholm C, Johnson K, et al. Extrafollicular dermal melanocyte stem cells and melanoma. Stem Cells Int. 2012;2012:407079.
  • 49
    Hoerter JD, Bradley P, Casillas A, Chambers D, Weiswasser B, Clements L, et al. Does Melanoma Begin in a Melanocyte Stem Cell? J Skin Cancer. 2012;2012:571087.
  • Financial Support: Maehara L de SN received a scholarship from CNPq (201591/2012-0)
  • How to cite this article: Ogliari KS, Marinowic D, Brum DE, Loth F. Stem cells in dermatology. An Bras Dermatol. 2014;89(2):286-91
  • *
    Work performed at Hemocord - Stem Cell Bank - Porto Alegre, RS, Brazil.

Publication Dates

  • Publication in this collection
    Mar-Apr 2014

History

  • Received
    11 Feb 2013
  • Accepted
    08 Apr 2013
Sociedade Brasileira de Dermatologia Av. Rio Branco, 39 18. and., 20090-003 Rio de Janeiro RJ, Tel./Fax: +55 21 2253-6747 - Rio de Janeiro - RJ - Brazil
E-mail: revista@sbd.org.br