151
|
Müller-Decker K, Manegold G, Butz H, Hinz DE, Hüttner D, Richter KH, Tremmel M, Weissflog R, Marks F. Inhibition of cell proliferation by bacterial lipopolysaccharides in TLR4-positive epithelial cells: independence of nitric oxide and cytokine release. J Invest Dermatol 2005; 124:553-61. [PMID: 15737196 DOI: 10.1111/j.0022-202x.2004.23598.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phylogenetically conserved toll-like receptors (TLR) recognize "pathogen-associated molecular patterns". Upon binding of ligands, TLR initiate innate immune response in immune and most likely epithelial cells. The TLR4 isoform is considered as a lipopolysaccharide (LPS) receptor. As shown here, a rat-tongue-derived epithelial cell line RTE2 expressed TLR4 mRNA and functional protein. LPS-treated RTE2 cells responded with the transient expression of inducible nitric oxide synthase (iNOS), an effector protein of TLR4 involved in the innate immune defense of monocytes. iNOS induction occurred along a nuclear factor-kappaB (NF-kappab)-dependent pathway and correlated with the increased production of NO. Moreover, LPS and lipid A were potent inhibitors of proliferation of RTE2 cells, of mouse keratinocytes, and mouse epidermis in vivo. The inhibition depended on lipid A structure, i.e., it was related to the endotoxin activity of LPS and at least in vitro was in part mediated by NF-kappaB. C57Bl/10 ScCr mice, lacking a functional TLR4, did not respond with growth inhibition, strongly suggesting a TLR4-mediated effect. RTE2 proliferation was also inhibited by transforming growth factor beta (TGFbeta) and tumor necrosis factor alpha (TNFalpha), whereas interferon gamma (IFNgamma) was a weak inhibitor. But the growth-inhibitory effect of LPS on RTE2 cells was not mediated by TNFalpha, TGFbeta, or NO. It is concluded that besides induction of innate immune responses, LPS specifically induces growth arrest in epithelial tongue cells and keratinocytes in vitro and in mouse epidermis in a TLR4-dependent but cytokine- and NO-independent manner.
Collapse
Affiliation(s)
- Karin Müller-Decker
- German Cancer Research Center, Research Project Eicosanoids and Tumor Development, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Mukhopadhyay A, Tan EKJ, Khoo YTA, Chan SY, Lim IJ, Phan TT. Conditioned medium from keloid keratinocyte/keloid fibroblast coculture induces contraction of fibroblast-populated collagen lattices. Br J Dermatol 2005; 152:639-45. [PMID: 15840092 DOI: 10.1111/j.1365-2133.2005.06545.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Keloid scars represent a pathological response to cutaneous injury. Overproliferation of fibroblasts and overproduction of collagen characterize these abnormal scars. The pathology of these scars remains poorly understood. The role of epithelial-mesenchymal interactions in keloid pathogenesis and scar contracture has recently been explored. OBJECTIVES To test our hypothesis that epithelial-mesenchymal interactions play a major role in modulating keloid scar contracture. METHODS A coculture model was employed wherein keloid and normal keratinocytes were cocultured with keloid or normal fibroblasts, and the conditioned media from day 5 cocultures were collected to study the effect of the paracrine secretions on contraction of an in vitro fibroblast-populated collagen lattice (FPCL) model. RESULTS Keloid keratinocyte/keloid fibroblast coculture conditioned media brought about increased contraction of the collagen lattice compared with non-cocultured conditioned media. When keloid fibroblasts populated the collagen lattice, significantly increased lattice contraction was induced compared with lattices populated by normal fibroblasts. The addition of antitransforming growth factor (TGF)-beta neutralizing antibody to the conditioned media produced an attenuation of the contraction of the FPCLs. When keloid and normal fibroblasts were cultured on chamber slides and treated with conditioned media from coculture and non-coculture series, immunohistochemical analysis demonstrated an increased expression of alpha-smooth muscle actin (a marker for fibroblast differentiation into myofibroblasts) in fibroblasts exposed to conditioned media from coculture. CONCLUSIONS These data indicate that epithelial-mesenchymal interactions are likely to play a major role in scar contracture and scar pathogenesis, and underscore the role of TGF-beta1 as a key player in keloid pathogenesis.
Collapse
Affiliation(s)
- A Mukhopadhyay
- Department of Surgery, The National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260
| | | | | | | | | | | |
Collapse
|
153
|
Lateef H, Abatan OI, Aslam MN, Stevens MJ, Varani J. Topical pretreatment of diabetic rats with all-trans retinoic acid improves healing of subsequently induced abrasion wounds. Diabetes 2005; 54:855-61. [PMID: 15734865 DOI: 10.2337/diabetes.54.3.855] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In the current study, rats were made diabetic with streptozotocin (STZ) and maintained for 8 weeks, during which time they were treated topically on alternative days with a solution of 0.1% all-trans retinoic acid in a vehicle of 70:30% ethanol/propylene glycol. STZ-induced diabetic rats treated with vehicle served as controls. Additional nondiabetic rats were treated with all-trans retinoic acid or vehicle in parallel. At the end of the 8-week period, rats from all four treatment groups were subjected to abrasion wound formation. Wounds healed more rapidly in vehicle-treated nondiabetic skin than in vehicle-treated diabetic skin (96% of the wound surface area closed in nondiabetic rats within 6 days vs. 41% closed in diabetic rats). Wounds in all-trans retinoic acid-treated diabetic skin healed more rapidly than wounds in vehicle-treated diabetic skin (85% of the wound surface area closed in all-trans retinoic acid-treated diabetic rats vs. 41% closed in vehicle-treated diabetic rats). At the histological level, recently healed skin from vehicle-treated diabetic rats was shown to contain a thin, wispy provisional matrix in which many of the embedded cells were rounded and some were pycnotic. In contrast, a much denser provisional matrix with large numbers of embedded spindle-shaped cells was observed in healed wounds from diabetic skin that had been pretreated with all-trans retinoic acid. The all-trans retinoic acid-treated diabetic skin was histologically similar to vehicle-treated (or all-trans retinoic acid-treated) skin from nondiabetic animals. In light of these findings, we suggest that prophylactic use of retinoid-containing preparations might be useful in preventing the development of nonhealing skin ulcers resultant from minor traumas in at-risk skin.
Collapse
Affiliation(s)
- Humaira Lateef
- Department of Pathology, University of Michigan, 1301 Catherine Road/Box 0602, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
154
|
Abstract
A major area of research in regenerative medicine is the potential application of stem cells in skin grafting and tissue engineering. This would require well defined and efficient protocols for directing the commitment and differentiation of stem cells into the keratinocyte lineage, together with their selective purification and proliferation in vitro. The development of such protocols would reduce the likelihood of spontaneous differentiation of stem cells into divergent lineages upon transplantation, as well as reduce the risk of teratoma formation in the case of embryonic stem cells. Additionally, such protocols could provide useful in vitro models for studying skin tissue biology, as well as facilitate the genetic manipulation of stem cells for therapeutic applications. The development of pharmacokinetic and cytotoxicity/genotoxicity screening tests for skin-related biomaterials and drugs could also utilize protocols developed for the commitment and differentiation of stem cells into the keratinocyte lineage. Hence, this review critically examines the various strategies that could be employed to direct the commitment and differentiation of stem cells into the keratinocyte lineage in vitro.
Collapse
Affiliation(s)
- Boon Chin Heng
- Stem Cell Laboratory, Faculty of Dentistry, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
155
|
Bellemare J, Roberge CJ, Bergeron D, Lopez-Vallé CA, Roy M, Moulin VJ. Epidermis promotes dermal fibrosis: role in the pathogenesis of hypertrophic scars. J Pathol 2005; 206:1-8. [PMID: 15772942 DOI: 10.1002/path.1737] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hypertrophic scarring is a pathological process characterized by fibroblastic hyperproliferation and by excessive deposition of extracellular matrix components. It has been hypothesized that abnormalities in epidermal-dermal crosstalk explain this pathology. To test this hypothesis, a tissue-engineered model of self-assembled reconstructed skin was used in this study to mimic interactions between dermal and epidermal cells in normal or pathological skin. These skin equivalents were constructed using three dermal cell types: normal wound (Wmyo) or hypertrophic wound (Hmyo) myofibroblasts and normal skin fibroblasts (Fb). Epidermis was reconstructed with normal skin keratinocytes (NK) or hypertrophic scar keratinocytes (HK). In the absence of keratinocytes, Hmyo formed a thicker dermis than Wmyo. When seeded with NK, the dermal thickness of Hmyo (121.2 +/- 31.4 microm vs 196.2 +/- 27.8 microm) and Fb (43.7 +/- 7.1 microm vs 83.6 +/- 16.3 microm) dermis was significantly (p < 0.05) reduced, while that of Wmyo (201.5 +/- 15.7 microm vs 160.7 +/- 21.1 microm) was increased. However, the presence of HK always induced significantly thicker dermis formation than observed with NK (Wmyo: 238.8 +/- 25.9 microm; Hmyo: 145.5 +/- 22.4 microm; Fb: 74.2 +/- 11.2 microm). These results correlated with collagen and MMP-1 secretion and with cell proliferation, which were increased when keratinocytes were added, except for the collagen secretion of Hmyo and Fb in the presence of NK. The level of dermal apoptosis was not different when epidermis was added to the dermis (<1% in each category). These observations strongly suggest that hypertrophic scar keratinocytes play a role in the development of pathological fibrosis by influencing the behaviour of dermal cells.
Collapse
Affiliation(s)
- Judith Bellemare
- Laboratoires des grands brûlés/LOEX, Hôpital du Saint-Sacrement du Centre Hospitalier Affilié Université de Québec, Québec, Canada
| | | | | | | | | | | |
Collapse
|
156
|
El Ghalbzouri A, Ponec M. Diffusible factors released by fibroblasts support epidermal morphogenesis and deposition of basement membrane components. Wound Repair Regen 2004; 12:359-67. [PMID: 15225215 DOI: 10.1111/j.1067-1927.2004.012306.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epithelial-mesenchymal interactions play an important role in controlling epidermal morphogenesis and homeostasis but little is known about the mechanisms of these interactions. To examine whether diffusible factors produced by fibroblasts and/or keratinocytes support epidermal morphogenesis and basement membrane formation, organotypic keratinocyte monocultures were established in media collected either from organotypic fibroblast or keratinocyte-monocultures or from keratinocyte-fibroblast cocultures, and the expression of keratin 10, 16, and 17 and basement membrane components (types IV and VII collagen, laminin 5, nidogen, BP 180, LAD-1) were examined. We found that diffusible factors released by keratinocytes were not sufficient to support the establishment of normalized epidermal phenotype and deposition of basement membrane components in contrast to fibroblast- or keratinocyte/fibroblast-derived factors. Keratinocytes appear to affect the spectrum of secreted soluble factors, as keratinocyte/fibroblast-derived factors were more effective to accomplish continuous linear deposition of laminin 5 and of nidogen. The finding that released amounts of keratinocyte growth factor and granulocyte macrophage colony stimulating factor were not sufficient to fully support epidermal morphogenesis and deposition of basement membrane components is suggestive for the involvement of other released diffusible factors. Generation of organotypic keratinocyte monocultures in the presence of fibroblast- or keratinocyte/fibroblast-derived soluble factors resulted in enhanced expression of keratins K16 and K17 and the absence of type IV collagen. This observation indicates that next to paracrine acting factors, epidermal homeostasis is controlled by mutual keratinocyte-fibroblast interaction.
Collapse
|
157
|
Finch PW, Rubin JS. Keratinocyte growth factor/fibroblast growth factor 7, a homeostatic factor with therapeutic potential for epithelial protection and repair. Adv Cancer Res 2004; 91:69-136. [PMID: 15327889 DOI: 10.1016/s0065-230x(04)91003-2] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Keratinocyte growth factor (KGF) is a paracrine-acting, epithelial mitogen produced by cells of mesenchymal origin. It is a member of the fibroblast growth factor (FGF) family, and acts exclusively through a subset of FGF receptor isoforms (FGFR2b) expressed predominantly by epithelial cells. The upregulation of KGF after epithelial injury suggested it had an important role in tissue repair. This hypothesis was reinforced by evidence that intestinal damage was worse and healing impaired in KGF null mice. Preclinical data from several animal models demonstrated that recombinant human KGF could enhance the regenerative capacity of epithelial tissues and protect them from a variety of toxic exposures. These beneficial effects are attributed to multiple mechanisms that collectively act to strengthen the integrity of the epithelial barrier, and include the stimulation of cell proliferation, migration, differentiation, survival, DNA repair, and induction of enzymes involved in the detoxification of reactive oxygen species. KGF is currently being evaluated in clinical trials to test its ability to ameliorate severe oral mucositis (OM) that results from cancer chemoradiotherapy. In a phase 3 trial involving patients who were treated with myeloablative chemoradiotherapy before autologous peripheral blood progenitor cell transplantation for hematologic malignancies, KGF significantly reduced both the incidence and duration of severe OM. Similar investigations are underway in patients being treated for solid tumors. On the basis of its success in ameliorating chemoradiotherapy-induced OM in humans and tissue damage in a variety of animal models, additional clinical applications of KGF are worthy of investigation.
Collapse
Affiliation(s)
- Paul W Finch
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
158
|
Marneros AG, Krieg T. Keloids - clinical diagnosis, pathogenesis, and treatment options. Keloide - klinische Diagnose, Pathogenese und Behandlungsoptionen. J Dtsch Dermatol Ges 2004; 2:905-13. [PMID: 16281608 DOI: 10.1046/j.1439-0353.2004.04077.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Keloids are defined as excessive scar tissue formation extending beyond the area of the original skin injury and occurring in predisposed individuals. They are considered to be a result of abnormal wound healing. The pathogenetic mechanisms that cause keloids remain unknown. Experiments with cells derived from keloid tissue revealed a number of abnormalities in cellular functions, such as in proliferation, apoptosis, or expression of growth factors and extracellular matrix proteins. Furthermore, several studies have reported altered keratinocyte-fibroblast interactions in keloids. Despite the diverse pathological changes in cellular functions and expression profiles of cells derived from keloid tissue, recent genetic studies have provided evidence that single genes may act as major regulators of keloid formation. We provide an overview of the pathogenetic mechanisms of keloid formation in the context of their clinical characteristics and current therapeutic approaches.
Collapse
|
159
|
|
160
|
Hobbs RM, Silva-Vargas V, Groves R, Watt FM. Expression of activated MEK1 in differentiating epidermal cells is sufficient to generate hyperproliferative and inflammatory skin lesions. J Invest Dermatol 2004; 123:503-15. [PMID: 15304090 DOI: 10.1111/j.0022-202x.2004.23225.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Epidermal activation of Erk MAPK is observed in human psoriatic lesions and in a mouse model of psoriasis in which beta1 integrins are expressed in the suprabasal epidermal layers. Constitutive activation of the upstream kinase MEK1 causes hyperproliferation and perturbed differentiation of human keratinocytes in culture. It is not known, however, whether Erk activation in differentiating keratinocytes is sufficient to trigger hyperproliferation of basal keratinocytes and a skin inflammatory infiltrate. To investigate this, we expressed constitutively active MEK1 in the suprabasal epidermal layers of transgenic mice. Proliferation in the epidermal basal layer was stimulated and epidermal terminal differentiation was perturbed. Some older mice also developed papillomas. There was a large increase in T lymphocytes, dendritic cells, and neutrophils in the skin. The effects of suprabasal MEK1 on basal keratinocytes and leukocytes, cells that were transgene negative, suggested that MEK1 activity might stimulate cytokine release. Transgenic keratinocytes expressed elevated IL-1alpha and crossing the mice with mice overexpressing the IL-1 receptor in the epidermal basal layer led to exacerbated hyperproliferation and inflammation. These data suggest that activation of MEK1 downstream of beta1 integrins plays an important role in epidermal hyperproliferation and skin inflammation.
Collapse
Affiliation(s)
- Robin M Hobbs
- Keratinocyte Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | | | | | | |
Collapse
|
161
|
Tudor D, Locke M, Owen-Jones E, Mackenzie IC. Intrinsic Patterns of Behavior of Epithelial Stem Cells. J Investig Dermatol Symp Proc 2004; 9:208-14. [PMID: 15369215 DOI: 10.1111/j.1087-0024.2004.09310.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The early concepts concerning hematopoietic and epithelial stem cells that were derived from kinetic studies have been greatly enhanced by new information about a range of other properties of somatic and embryonic stem cells. Firstly, the stem and amplifying pattern characteristically established by epithelial lineages has been found to represent an intrinsic pattern that is generated by somatic epithelial stem cells without the need for additional environmental information. Secondly, it is now apparent that somatic epithelial stem cells are plastic and can be directed into a range of new pathways of differentiation by heterotypic interactions. The mechanisms of this plasticity need to be reconciled with the normally stable commitment of these cells to production only of progeny entering a tightly restricted range of phenotypic pathways. The present review discusses the intrinsic properties of epithelial stem cells and how they may be acted upon by connective tissues to generate a wide range of phenotypically different epithelial structures.
Collapse
Affiliation(s)
- Debbie Tudor
- University of Wales, College of Medicine, Heath Park, Cardiff, UK
| | | | | | | |
Collapse
|
162
|
Kampinga HH, Van Waarde-Verhagen MAWH, Van Assen-Bolt AJ, Nieuwenhuis B, Rodemann HP, Prowse KR, Linskens MHK. Reconstitution of active telomerase in primary human foreskin fibroblasts: effects on proliferative characteristics and response to ionizing radiation. Int J Radiat Biol 2004; 80:377-88. [PMID: 15223771 DOI: 10.1080/09553000410001692735] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Telomere shortening has been proposed to trigger senescence, and since most primary cells do not express active telomerase, reactivation of telomerase activity was proposed as a safe and non-transforming way of immortalizing cells. However, to study radiation responses, it is as yet unclear whether cells immortalized by telomerase reactivation behave in a similar manner as their parental primary cells. MATERIALS AND METHODS Primary human foreskin fibroblasts were transfected with the human catalytic subunit of telomerase, the reverse transcriptase (hTERT), and their growth characteristics and response to DNA damage were characterized. RESULTS The sole expression of the human hTERT was sufficient to immortalize the human foreskin fibroblasts. With time in culture, the immortalized cells almost doubled their average telomeric length and the clonal population contained almost no post-mitotic fibroblasts anymore. Up to 300 population doublings, no alterations compared with the parental primary cells were seen in terms of clonogenic radiosensitivity, DNA double-strand break repair, radiation-induced increases in p53 and p21(WAF-1,CIP-1) expression, and the G1/S and G2/M cell cycle checkpoints. Moreover, mitogen-induced mitotic arrest of fibroblasts was still possible in the hTERT-immortalized clones. CONCLUSIONS Immortalizing fibroblasts by reconstitution of active telomerase seems a good, reliable manner to generate a large source of cells with a radiation damage response similar to the primary cells.
Collapse
Affiliation(s)
- H H Kampinga
- Department of Radiation and Stress Cell Biology, DCB, Faculty of Medical Sciences, University of Groningen, Groningen 9713 AV, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
163
|
Shephard P, Martin G, Smola-Hess S, Brunner G, Krieg T, Smola H. Myofibroblast differentiation is induced in keratinocyte-fibroblast co-cultures and is antagonistically regulated by endogenous transforming growth factor-beta and interleukin-1. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:2055-66. [PMID: 15161640 PMCID: PMC1615767 DOI: 10.1016/s0002-9440(10)63764-9] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In wound healing epidermal-dermal interactions are known to regulate keratinocyte proliferation and differentiation. To find out how fibroblasts respond to epithelial stimuli, we characterized fibroblasts in monolayer co-culture with keratinocytes. On co-culture numerous extracellular matrix- and smooth muscle cell-associated gene transcripts were up-regulated in fibroblasts, suggesting a differentiation into myofibroblasts. Increased alpha-smooth muscle actin (alpha-SMA) protein expression in co-cultured fibroblasts started at approximately day 4, was serum-independent, but required endogenous transforming growth factor (TGF)-beta. In co-cultures, TGF-beta neutralizing monoclonal antibody strongly reduced alpha-SMA induction. Endogenous TGF-beta production and activation were increased at 24 and 48 hours, requiring, like alpha-SMA induction, close keratinocyte-fibroblast proximity. As myofibroblast differentiation only started after 4 days, we analyzed the presence of endogenous inhibitors at early time points. Blocking keratinocyte-derived interleukin (IL)-1 using IL-1 receptor antagonist, alpha-SMA expression in co-cultures was potentiated. Conversely, adding exogenous IL-1alpha completely suppressed endogenous alpha-SMA induction. In co-cultured fibroblasts strong nuclear factor-kappaB binding activity was observed from 2 hours, decreasing at 2 and 4 days, suggesting an early, IL-1-mediated inhibition of TGF-beta signaling in co-cultured fibroblasts. This biphasic differentiation event is regulated by the balance of endogenous TGF-beta and IL-1 activity and is reminiscent of myofibroblast differentiation at early and later stages of wound healing.
Collapse
Affiliation(s)
- Pierre Shephard
- Departments of Dermatology and Virology, University of Cologne, Cologne, Germany
| | | | | | | | | | | |
Collapse
|
164
|
Sorrell JM, Baber MA, Caplan AI. Site-matched papillary and reticular human dermal fibroblasts differ in their release of specific growth factors/cytokines and in their interaction with keratinocytes. J Cell Physiol 2004; 200:134-45. [PMID: 15137066 DOI: 10.1002/jcp.10474] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The interfollicular dermis of adult human skin is partitioned into histologically and physiologically distinct papillary and reticular zones. Each of these zones contains a unique population of fibroblasts that differ in respect to their proliferation kinetics, rates at which they contract type I collagen gels, and in their relative production of decorin and versican. Here, site-matched papillary and reticular dermal fibroblasts couples were compared to determine whether each population interacted with keratinocytes in an equivalent or different manner. Papillary and reticular fibroblasts grown in monolayer culture differed significantly from each other in their release of keratinocyte growth factor (KGF) and granulocyte-macrophage colony stimulating factor (GM-CSF) into culture medium. Some matched fibroblast couples also differed in their constitutive release of interleukin-6 (IL-6). Papillary fibroblasts produced a higher ratio of GM-CSF to KGF than did corresponding reticular fibroblasts. Interactions between site-matched papillary and reticular couples were also assayed in a three-dimensional culture system where fibroblasts and keratinocytes were randomly mixed, incorporated into type I collagen gels, and allowed to sort. Keratinocytes formed distinctive cellular masses in which the keratinocytes were organized such that the exterior most layer of cells exhibited characteristics of basal keratinocytes and the interior most cells exhibited characteristics of terminally differentiated keratinocytes. In the presence of papillary dermal fibroblasts, keratinocyte masses were highly symmetrical and cells expressed all levels of differentiation markers. In contrast, keratinocyte masses that formed in the presence of reticular fibroblasts tended to have irregular shapes, and terminal differentiation was suppressed. Furthermore, basement membrane formation was retarded in the presence of reticular cells. These studies indicate that site-matched papillary and reticular dermal fibroblasts qualitatively differ in their support of epidermal cells, with papillary cells interacting more effectively than corresponding reticular cells.
Collapse
Affiliation(s)
- J Michael Sorrell
- Biology Department, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | | | | |
Collapse
|
165
|
Abstract
Dermal fibroblasts are a dynamic and diverse population of cells whose functions in skin in many respects remain unknown. Normal adult human skin contains at least three distinct subpopulations of fibroblasts, which occupy unique niches in the dermis. Fibroblasts from each of these niches exhibit distinctive differences when cultured separately. Specific differences in fibroblast physiology are evident in papillary dermal fibroblasts, which reside in the superficial dermis, and reticular fibroblasts, which reside in the deep dermis. Both of these subpopulations of fibroblasts differ from the fibroblasts that are associated with hair follicles. Fibroblasts engage in fibroblast-epidermal interactions during hair development and in interfollicular regions of skin. They also play an important role in cutaneous wound repair and an ever-increasing role in bioengineering of skin. Bioengineered skin currently performs important roles in providing (1) a basic understanding of skin biology, (2) a vehicle for testing topically applied products and (3) a resource for skin replacement.
Collapse
Affiliation(s)
- J Michael Sorrell
- Skeletal Research Center, Biology Department, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | |
Collapse
|
166
|
Nowinski D, Lysheden AS, Gardner H, Rubin K, Gerdin B, Ivarsson M. Analysis of Gene Expression in Fibroblasts in Response to Keratinocyte-Derived Factors In Vitro: Potential Implications for the Wound Healing Process11Table 1, Table 2 and Table 5 can be found online at http://www.blackwellpublishing.com/products/journals/suppmat/jid/jid22112/jid22112sm.htm. J Invest Dermatol 2004; 122:216-21. [PMID: 14962111 DOI: 10.1046/j.0022-202x.2003.22112.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cocultures of fibroblasts and keratinocytes, physically separated by a membrane, were carried out for 48 h, and large-scale gene expression in the fibroblasts was analyzed by Affymetrix microarrays of expressed mRNAs. Two independent experiments were performed with cells from different individuals. A total of 243 genes were upregulated twofold or more and 100 genes were negatively regulated (reduction by half or more) in both experiments. A total of 69 of these 343 genes coded for growth factors, cytokines, chemokines, or their receptors; extracellular matrix molecules or enzymes involved in their synthesis; adhesion receptors; proteinases/proteinase inhibitors or their receptors; cell cycle regulators; apoptosis-regulating factors; prostaglandin-related factors; or growth-factor-binding proteins. Most of the remaining genes coded for proteins involved in signal transduction or general metabolism. Sixteen genes selected from these groups were further analyzed by northern blot analysis to confirm the array data. Finally, experiments with interleukin-1 alpha (IL-1 alpha)-blocking antibodies or IL-1 receptor antagonists demonstrate that IL-1 alpha is one important factor involved in keratinocyte-mediated regulation of gene expression in fibroblasts. Taken together, the results suggest that keratinocytes regulate fibroblast gene expression with implications for the wound healing process during reepithelialization.
Collapse
Affiliation(s)
- Daniel Nowinski
- Department of Surgical Sciences, Plastic Surgery Unit, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
167
|
El Ghalbzouri A, Hensbergen P, Gibbs S, Kempenaar J, van der Schors R, Ponec M. Fibroblasts facilitate re-epithelialization in wounded human skin equivalents. J Transl Med 2004; 84:102-12. [PMID: 14631386 DOI: 10.1038/labinvest.3700014] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The re-epithelialization of the wound involves the migration of keratinocytes from the edges of the wound. During this process, keratinocyte migration and proliferation will depend on the interaction of keratinocytes with dermal fibroblasts and the extracellular matrix. The present study aimed to investigate (1) the role of fibroblasts in the re-epithelialization process and on the reconstitution of the dermal-epidermal junction (DEJ) and (2) differential protein expression during re-epithelialization. For both purposes, three-dimensional human skin equivalents (HSE) were used. A full-thickness wound in HSE was introduced by freezing with liquid nitrogen and a superficial wound by linear incision with a scalpel. The closure of the wound in the absence or presence of exogenous growth factors was followed by monitoring the rate of re-epithelialization and regeneration of the DEJ. The results obtained in this study demonstrate that fibroblasts facilitate wound closure, but they differentially affected the deposition of various basement membrane components. The deposition of laminin 5 at the DEJ was delayed in superficial wounds as compared to the full-thickness wounds. During freeze injury, some basement membrane (BM) components remain associated with the dermal compartment and probably facilitate the BM reconstitution. The re-epithelialization process in full-thickness but not in superficial wounds was accelerated by the presence of keratinocyte growth factor and especially by epidermal growth factor. In addition, we have examined the deposition of various basement membrane components and the differences in protein expression in a laterally expanding epidermis in uninjured HSE. Laminin 5, type IV and VII collagen deposition was decreased in the laterally expanding epidermis, indicating that the presence of these proteins is not required for keratinocyte migration to occur in vitro. Using two-dimensional polyacrylamide gel electrophoresis, we have identified DJ-1, a protein not earlier reported to be differently expressed during the epithelialization process of the skin.
Collapse
|
168
|
El Ghalbzouri A, Jonkman M, Kempenaar J, Ponec M. Recessive epidermolysis bullosa simplex phenotype reproduced in vitro: ablation of keratin 14 is partially compensated by keratin 17. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:1771-9. [PMID: 14578178 DOI: 10.1016/s0002-9440(10)63537-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Recessive epidermolysis bullosa simplex (REBS) is characterized by generalized cutaneous blistering in response to mechanical trauma. This results from fragility of the basal keratinocytes that lack keratin tonofilaments because of homozygote null mutation in the keratin 14 gene. REBS patients display in addition focal dyskeratotic skin lesions with histology of epidermolytic hyperkeratosis (EHK) and tonofilament clumping in the suprabasal layers of the epidermis. In this study we examined whether it is possible to mimic in vitro the bullous and dyskeratotic cellular phenotype. For this purpose, fibroblasts from nondyskeratotic (K14-/-) and dyskeratotic (K14-/-) skin of a REBS patient and fibroblasts from a healthy donor (K14+/+) were isolated and incorporated into collagen matrices. Subsequently, fresh biopsies originating from the nondyskeratotic and dyskeratotic skin of the patient and from a healthy donor were placed onto the collagen matrices and cultured at the air-liquid interface. Epidermal morphogenesis was evaluated on the basis of tissue morphology and the expression of a series of keratins. The results of the present study indicate that basal cell vacuolization in REBS can be mimicked in vitro but not the EHK. Fibroblasts seem to play an important regulatory role in establishing the REBS phenotype. These findings suggest that wild-type fibroblasts may enhance the stability of K14-/- keratinocytes in vitro.
Collapse
Affiliation(s)
- Abdoelwaheb El Ghalbzouri
- Department of Dermatology, Leiden University Medical Center, Sylvius Laboratory, 2300 RA Leiden, The Netherlands
| | | | | | | |
Collapse
|
169
|
Funayama E, Chodon T, Oyama A, Sugihara T. Keratinocytes Promote Proliferation and Inhibit Apoptosis of the Underlying Fibroblasts: An Important Role in the Pathogenesis of Keloid. J Invest Dermatol 2003; 121:1326-31. [PMID: 14675177 DOI: 10.1111/j.1523-1747.2003.12572.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interactions between epidermal keratinocytes and dermal fibroblasts play an important role in regulating tissue homeostasis and repair. Nevertheless, little is known about the role of keratinocytes in the pathogenesis of keloid. In this study, we investigated the influence of normal skin- and keloid-derived keratinocytes on normal skin- and keloid-derived fibroblasts utilizing a serum-free indirect coculture system. The keloid-derived fibroblasts showed a greater proliferation and minimal apoptosis when cocultured with normal skin- or keloid-derived keratinocytes, and the results were most significant in the latter. This difference was not observed when the fibroblasts were treated with conditioned medium obtained from normal skin- and keloid-derived keratinocytes. Nevertheless, conditioned medium-treated groups showed more proliferation and less apoptosis compared to the nonconditioned medium-treated control groups. We also analyzed the profile of factors involved in cell growth and apoptosis in fibroblasts cocultured with keratinocytes. Extracellular signal-regulated kinase and c-Jun N-terminal kinase phosphorylations and expression of Bcl-2 and transforming growth factor-beta1 were all significantly upregulated in the fibroblasts cocultured with keloid-derived keratinocytes. Together, these results strongly suggest that the overlying keratinocytes of the keloid lesion play an important role in keloidogenesis by promoting more proliferation and less apoptosis in the underlying fibroblasts through paracrine and double paracrine effects.
Collapse
Affiliation(s)
- Emi Funayama
- Department of Plastic and Reconstructive Surgery, Hokkaido University School of Medicine, Sapporo, Japan
| | | | | | | |
Collapse
|
170
|
Costea DE, Loro LL, Dimba EAO, Vintermyr OK, Johannessen AC. Crucial Effects of Fibroblasts and Keratinocyte Growth Factor on Morphogenesis of Reconstituted Human Oral Epithelium. J Invest Dermatol 2003; 121:1479-86. [PMID: 14675199 DOI: 10.1111/j.1523-1747.2003.12616.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The connective tissue is known to have a general supportive effect for the development of the overlying epithelium; however, the more specific effects of fibroblasts and the involvement of their product, keratinocyte growth factor, on oral epithelial morphogenesis have not yet been addressed. Therefore, the purpose of this study was to investigate the effects of fibroblasts and keratinocyte growth factor on human oral epithelial morphogenesis in vitro. Reconstituted human oral epithelium was generated from primary human oral keratinocytes and fibroblasts by use of an organotypic cell culture model in a defined medium. Addition of fibroblasts to the collagen biomatrix increased total epithelial thickness from 28.0+/-5.0 microm to 66.1+/-8.6 microm (p=0.028), and basal cell proliferation from 3.6+/-0.7% to 16.6+/-1.1% (p=0.025). Presence of fibroblasts profoundly influenced the pattern of epithelial differentiation, and induced a switch in the pattern of cell death, from a predominance of spontaneous cell death in the basal cell layer (from 4.7+/-0.6% to 1.8+/-0.3%, p=0.029) to a more prevalent cell death due to terminal differentiation in the suprabasal cell layer (from 4.0+/- 0.1% to 5.4+/-0.1%, p=0.034). Keratinocyte growth factor promoted epithelial growth, but did not significantly enhance epithelial differentiation, demonstrating that fibroblasts possess additional mechanisms to keratinocyte growth factor synthesis that can modulate differentiation of reconstituted human oral epithelium.
Collapse
Affiliation(s)
- Daniela Elena Costea
- Department of Odontology-Oral Pathology and Forensic Odontology, Faculty of Dentistry, University of Bergen, Norway
| | | | | | | | | |
Collapse
|
171
|
Yang GP, Lim IJ, Phan TT, Lorenz HP, Longaker MT. From scarless fetal wounds to keloids: Molecular studies in wound healing. Wound Repair Regen 2003; 11:411-8. [PMID: 14617279 DOI: 10.1046/j.1524-475x.2003.11604.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Surgical researchers were among the first to describe the different phases of wound healing and the events in tissue repair and regeneration that were taking place during each phase. The understanding of these events has been significantly enhanced in recent years by modern techniques in molecular and cellular biology. In this article, we discuss new findings in scarless fetal repair, angiogenesis in wound healing, and keloid pathogenesis. This serves to highlight the advances that have been made and also how much remains to be understood.
Collapse
Affiliation(s)
- George P Yang
- Department of Surgery, Stanford University, Stanford, California 94305, USA.
| | | | | | | | | |
Collapse
|
172
|
Lim IJ, Phan TT, Tan EK, Nguyen TTT, Tran E, Longaker MT, Song C, Lee ST, Huynh HT. Synchronous activation of ERK and phosphatidylinositol 3-kinase pathways is required for collagen and extracellular matrix production in keloids. J Biol Chem 2003; 278:40851-8. [PMID: 12907681 DOI: 10.1074/jbc.m305759200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Keloid fibroproliferation appears to be influenced by epithelial-mesenchymal interactions between keloid keratinocytes (KKs) and keloid fibroblasts (KFs). Keloid and normal fibroblasts exhibit accelerated proliferation and collagen I and III production in co-culture with KKs compared with single cell culture or co-culture with normal keratinocytes. ERK and phosphatidylinositol 3-kinase (PI3K) pathway activation has been observed in excessively proliferating KFs in co-culture with KKs. We hypothesized that ERK and PI3K pathways might be involved in collagen and extracellular matrix production in KFs. To test our hypothesis, four samples of KFs were co-cultured in defined serum-free medium with KKs for 2-5 days. KF cell lysate was subjected to Western blot analysis. Compared with KF single cell culture, phospho-ERK1/2 and downstream phospho-Elk-1 showed up-regulation in the co-culture groups, as did phospho-PI3K and phospho-Akt-1, indicating ERK and PI3K pathway activation. Western blotting of the conditioned medium demonstrated increased collagen I-III, laminin beta2, and fibronectin levels. Addition of the MEK1/2-specific inhibitor U0126 or the PI3K-specific inhibitor LY294002 (but not p38 kinase and JNK inhibitors) completely nullified collagen I-III production and significantly decreased laminin beta2 and fibronectin secretion. In the presence of the MEK1/2 or PI3K inhibitor, fibronectin demonstrated changes in molecular mass reflected by faster in-gel migration. These data strongly suggest that synchronous activation of both the ERK and PI3K pathways is essential for collagen I-III and laminin beta2 production. These pathways additionally appear to affect the side chain attachments of fibronectin. Modulation of these pathways may suggest a direction for keloid therapy.
Collapse
Affiliation(s)
- Ivor J Lim
- Departments of Surgery and Orthopaedic Surgery, National University of Singapore, 5, Lower Kent Ridge Road, Singapore 119074
| | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Marchese C, Visco V, Aimati L, Cardinali G, Kovacs D, Buttari B, Bellocci M, Torrisi MR, Picardo M. Nickel-induced keratinocyte proliferation and up-modulation of the keratinocyte growth factor receptor expression. Exp Dermatol 2003; 12:497-505. [PMID: 12930308 DOI: 10.1034/j.1600-0625.2002.120419.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Keratinocytes play a key role in the pathogenesis of allergic contact dermatitis (ADC) induced by the sensitizing agent nickel. We analyzed here the effects of treatment with nickel and of the pretreatment with zinc on HaCaT cells and primary human keratinocytes. Cell counting, 5-bromo-2'-deoxyuridine incorporation assay and adenosine triphosphate (ATP) bioluminescence detection showed that treatment with NiSO4 induced DNA synthesis and cell proliferation and that pretreatment with ZnSO4 was able to abrogate this proliferative effect. This nickel-induced cell growth appeared enhanced when primary human keratinocytes were co-cultured with fibroblasts. Western blot analysis demonstrated that nickel ions induced up-modulation of the expression of the keratinocyte growth factor receptors (KGFR) without affecting the keratinocyte differentiation, whereas the protein levels of the epidermal growth factor receptor (EGFR) and of its ligand transforming growth factor-alpha (TGF-alpha) appeared unmodified by the treatment. Double immunofluorescence showed that the effect of nickel on DNA synthesis was mainly exerted on KGFR expressing cells, suggesting that KGFR up-modulation could be required for the nickel-induced cell proliferation. These results indicate that KGFR and its ligands may play a role in the mechanism of action of nickel ions and in the protective effect of zinc pretreatment.
Collapse
Affiliation(s)
- Cinzia Marchese
- Dipartimento di Medicina Sperimentale e Patologia, Università di Roma 'La Sapienza'; Istituto Dermatologico San Gallicano, Roma, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Maas-Szabowski N, Stärker A, Fusenig NE. Epidermal tissue regeneration and stromal interaction in HaCaT cells is initiated by TGF-alpha. J Cell Sci 2003; 116:2937-48. [PMID: 12771184 DOI: 10.1242/jcs.00474] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The human keratinocyte cell line HaCaT expresses essentially all epidermal differentiation markers but exhibits deficiencies in tissue organization as surface transplants in nude mice and even more so in organotypic co-cultures with fibroblasts. Whereas tissue differentiation by normal keratinocytes (NEKs) is regulated by stromal interactions, this mechanism is impaired in HaCaT cells. This regulatory process is initiated by interleukin-1 (IL-1) release in keratinocytes, which induces expression of keratinocyte growth factor (KGF/FGF-7) and granulocyte macrophage-colony stimulating factor (GM-CSF) in fibroblasts. Production and release of IL-1 is very low and, consequently, expression of the fibroblast-derived growth factors KGF/FGF-7 and GM-CSF is absent in HaCaT-fibroblast co-cultures. However, addition of KGF and GMCSF, respectively, is inefficient to improve stratification and differentiation by HaCaT cells due to the low expression of their cognate receptors. More importantly, expression and release of the autocrine keratinocyte growth factor TGF-alpha is dramatically decreased in HaCaT cells. Addition of TGF- alpha or EGF stimulated HaCaT cell proliferation but, even more effectively, suppressed apoptosis, thus facilitating the formation of a regularly stratified epithelium. Furthermore, TGF-alpha enhanced the expression of the receptors for KGF and GM-CSF so that addition of these growth factors, or of their inducer IL-1, further improved epidermal tissue differentiation leading to in vitro skin equivalents comparable with cultures of NEKs. Thus, supplementing TGF-alpha normalized epidermal tissue regeneration by immortal HaCaT keratinocytes and their interaction with stromal cells so that regular skin equivalents are produced as standardized in vitro models.
Collapse
Affiliation(s)
- Nicole Maas-Szabowski
- Division of Differentiation and Carcinogenesis (B0600), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
175
|
Phan TT, Lim IJ, Bay BH, Qi R, Longaker MT, Lee ST, Huynh H. Role of IGF system of mitogens in the induction of fibroblast proliferation by keloid-derived keratinocytes in vitro. Am J Physiol Cell Physiol 2003; 284:C860-9. [PMID: 12620890 DOI: 10.1152/ajpcell.00350.2002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Keloids are proliferative dermal growths representing a pathological wound-healing response. We report high proliferation rates in normal (NF) and keloid-derived fibroblasts (KF) cocultured with keloid-derived keratinocytes (KK). IGF binding protein (IGFBP)-3 mRNA and secreted IGFBP-3 in conditioned media were increased in NF cocultured with KK compared with NF but markedly reduced in KF cocultured with KK or normal keratinocytes (NK). IGFBP-2 and IGFBP-4 mRNA levels were elevated, whereas IGFBP-5 mRNA was decreased in KF cocultured with KK or NK. Significant increases in IGFBP-2 and -4 mRNA in KF cocultured with KK did not correlate with protein secretion. Downstream IGF signaling cascade components, phospho-Raf, phospho-MEK1/2, phospho-MAPK, PI-3 kinase, phospho-Akt, and phospho-Elk-1, were elevated in KF cocultured with KK. Addition of recombinant human IGFBP-3 or antibodies against IGF-I or IGF-IR significantly inhibited proliferation of KF. The bioavailability of IGF-I may be related to the levels of IGFBP-3 produced, which in turn influences KF proliferation, suggesting that modulation of IGF-I, IGF-IR, and IGFBP-3, individually or in combination, may represent novel approaches to the treatment of keloids.
Collapse
Affiliation(s)
- Toan-Thang Phan
- National Burns Centre, Singapore General Hospital, Singapore 169608
| | | | | | | | | | | | | |
Collapse
|
176
|
Limat A, French LE, Blal L, Saurat JH, Hunziker T, Salomon D. Organotypic cultures of autologous hair follicle keratinocytes for the treatment of recurrent leg ulcers. J Am Acad Dermatol 2003; 48:207-14. [PMID: 12582390 DOI: 10.1067/mjd.2003.69] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Our purpose was to evaluate, in an open study, the efficacy of epidermal equivalents (EEs), a tissue-engineered epidermis prepared from autologous hair follicle keratinocytes, for the treatment of recurrent leg ulcers (n = 50). To generate EEs, keratinocytes expanded from the outer root sheaths of plucked anagen hair follicles were seeded on cell culture inserts at air-liquid interface. The total culture time was 5 to 6 weeks. Three days after the procedure, 95% of EEs adhered to the wound bed. After 8 weeks, 70% of the total wound surface was re-epithelialized and 32% of the ulcers were healed. After applying the EEs, a major relief of wound pain was noticed by the patients. EEs were applied in ambulatory patients without surgical facilities. Because 92% of the cases included in this study presented a recurrence of their ulcers after a split-thickness skin graft, we consider these ulcers as difficult to treat and propose the EEs as an alternative effective treatment of recurrent leg ulcers.
Collapse
Affiliation(s)
- Alain Limat
- Clinic of Dermatology, Hôpital Cantonal, University Hospital Geneva, HUG et DHURDV, 24 rue Micheli-du-Crest, CH-1211 Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
177
|
Grøn B, Stoltze K, Andersson A, Dabelsteen E. Oral fibroblasts produce more HGF and KGF than skin fibroblasts in response to co-culture with keratinocytes. APMIS 2002; 110:892-8. [PMID: 12645668 DOI: 10.1034/j.1600-0463.2002.1101208.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The production of hepatocyte growth factor (HGF) and keratinocyte growth factor (KGF) in subepithelial fibroblasts from buccal mucosa, periodontal ligament, and skin was determined after co-culture with keratinocytes. The purpose was to detect differences between the fibroblast subpopulations that could explain regional variation in epithelial growth and wound healing. Normal human fibroblasts were cultured on polystyrene or maintained in collagen matrix and stimulated with keratinocytes cultured on membranes. The amount of HGF and KGF protein in the culture medium was determined every 24 h for 5 days by ELISA. When cultured on polystyrene, the constitutive level of KGF and HGF in periodontal fibroblasts was higher than the level in buccal and skin fibroblasts. In the presence of keratinocytes, all three types of fibroblasts in general increased their HGF and KGF production 2-3 times. When cells were maintained in collagen, the level of HGF and KGF was decreased mainly in skin cultures. However, in oral fibroblasts, induction after stimulation was at a similar level in collagen compared to on polystyrene. Skin fibroblasts maintained in collagen produced almost no HGF whether with or without stimulation. The results demonstrate that the secretion of KGF and HGF in both unstimulated fibroblasts and in fibroblasts co-cultured with keratinocytes is dependent on the type of fibroblasts. In general, the periodontal fibroblasts had the highest level of cytokine production. This high level of growth factor production may influence the proliferation and the migration of junctional epithelium and thereby influence the development of periodontal disease.
Collapse
Affiliation(s)
- Birgitte Grøn
- Bartholin Institute, Kommunehospitalet, Copenhagen, Denmark
| | | | | | | |
Collapse
|
178
|
Ninomiya T, Kubota Y, Koji T, Shirasuna K. Marsupialization inhibits interleukin-1α expression and epithelial cell proliferation in odontogenic keratocysts. J Oral Pathol Med 2002; 31:526-33. [PMID: 12269991 DOI: 10.1034/j.1600-0714.2002.00029.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Marsupialization results in the reduction of odontogenic cyst size. Interleukin-1alpha (IL-1alpha) is thought to play a crucial role for the expansion of odontogenic keratocysts. The aim of this study was to investigate the effects of marsupialization on the expression of IL-1alpha and on the proliferating activity of a lining epithelium in odontogenic keratocysts. METHODS The concentrations of IL-1alpha, interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) in the intracystic fluids of odontogenic keratocysts were measured by the enzyme-linked immunosorbent assay (ELISA). The expression of IL-1alpha mRNA in odontogenic keratocysts was measured before and after marsupialization by in situ hybridization. The expression of IL-1alpha and epithelial cell-proliferating activities in odontogenic keratocysts were also measured by immunohistochemistry using antibodies for human IL-1alpha and Ki-67 antigen, respectively. RESULTS The intracystic fluid levels of IL-1alpha were significantly higher than those of IL-6 and TNF-alpha in odontogenic keratocysts. In situ hybridization and immunohistochemistry showed that strong expression of IL-1alpha mRNA and protein was mainly detected in the epithelial cells of odontogenic keratocysts. After marsupialization, the signal intensities for IL-1alpha mRNA and protein were significantly decreased. In addition, the Ki-67 labeling index of the epithelial cells was decreased proportionally with the grade of IL-1alpha mRNA expression after the marsupialization. CONCLUSION Our findings suggest that marsupialization may reduce the size of odontogenic keratocyst by inhibiting IL-1alpha expression and the epithelial cell proliferation.
Collapse
Affiliation(s)
- Tomohiro Ninomiya
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | |
Collapse
|
179
|
Angel P, Szabowski A. Function of AP-1 target genes in mesenchymal-epithelial cross-talk in skin. Biochem Pharmacol 2002; 64:949-56. [PMID: 12213591 DOI: 10.1016/s0006-2952(02)01158-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An increasing number of examples on the importance of mesenchymal-epithelial interactions in physiological (e.g. embryonic development) and pathological (tumourigenesis) processes have been described. This is best illustrated in the skin, where the well-controlled balance of keratinocyte proliferation and differentiation forms the basis for a proper histoarchitecture of the epidermis. Here, a double paracrine loop of cytokines, which are synthesised and secreted by cells of the epidermis (keratinocytes) and the underlying dermis (fibroblasts) seems to play a major role. The aim of this commentary is to review research that has investigated the role of specific subunits of transcription factor AP-1 (Jun/Fos) in this regulatory network. Using an in vitro skin equivalent model strong evidence was provided for a critical and specific function of c-Jun and JunB in mesenchymal-epithelial interaction in the skin by regulating the expression of interleukin-1 (IL-1)-induced keratinocyte growth factor (KGF) and GM-CSF in fibroblasts. These factors, in turn, adjust the balance between proliferation and differentiation of keratinocytes ensuring proper architecture of the epidermis. This commentary will summarise our current knowledge on the molecular mechanisms underlying AP-1-dependent mesenchymal-epithelial interactions and discuss the physiological relevance of these in vitro findings in skin physiology and pathology.
Collapse
Affiliation(s)
- Peter Angel
- Deutsches Krebsforschungszentrum, Division of Signal Transduction and Growth Control, Im Neuenheimer Feld 280, Heidelberg, Germany.
| | | |
Collapse
|
180
|
Moon SE, Bhagavathula N, Varani J. Keratinocyte stimulation of matrix metalloproteinase-1 production and proliferation in fibroblasts: regulation through mitogen-activated protein kinase signalling events. Br J Cancer 2002; 87:457-64. [PMID: 12177784 PMCID: PMC2376127 DOI: 10.1038/sj.bjc.6600478] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2001] [Revised: 05/10/2002] [Accepted: 05/16/2002] [Indexed: 11/19/2022] Open
Abstract
Incubation of human dermal fibroblasts in keratinocyte-conditioned culture medium led to a 5.7-fold increase in the level of matrix metalloproteinase-1. Virtually all of the matrix metalloproteinase-1 - inducing activity could be related to agonists acting through members of the epidermal growth factor receptor family or to agonists acting through the interleukin-1 receptor. The same keratinocyte-conditioned medium also induced a modest increase in fibroblast proliferation (approximately 1.8-fold). Growth-stimulating activity could be attributed to epidermal growth factor receptor (but not interleukin-1 receptor) function. In fibroblasts exposed to keratinocyte-conditioned medium, mitogen-activated protein kinase signalling through both the extracellular signal-related kinase pathway and p38 pathway occurred. When recombinant epidermal growth factor or recombinant interleukin-1beta were used as a control, they induced mitogen-activated protein kinase signalling consistent with the combined effects of epidermal growth factor receptor - specific and interleukin-1 receptor - specific agonists in keratinocyte-conditioned medium. Recombinant epidermal growth factor stimulated both matrix metalloproteinase-1 induction and proliferation while recombinant interleukin-1beta stimulated matrix metalloproteinase-1 elaboration but not fibroblast growth. An inhibitor of extracellular signal-related kinase pathway signalling (U0126) blocked induction of matrix metalloproteinase-1 production induced by keratinocyte-conditioned medium (as well as by epidermal growth factor or interleukin-1beta), and also inhibited proliferation. A p38 signalling inhibitor (SB203580) blocked matrix metalloproteinase-1 elaboration induced by keratinocyte-conditioned medium or interleukin-1beta, but did not inhibit matrix metalloproteinase-1 elaboration or cell growth induced by epidermal growth factor. These data indicate that keratinocyte-fibroblast interactions are mediated by multiple stimulating agents acting on specific receptors to induce signalling through different mitogen-activated protein kinase pathways leading to altered expression of key biological functions.
Collapse
Affiliation(s)
- S E Moon
- Department of Pathology, The University of Michigan Medical School, 1301 Catherine Road, Box 0602, Ann Arbor, Michigan, MI 48109, USA
| | | | | |
Collapse
|
181
|
el-Ghalbzouri A, Gibbs S, Lamme E, Van Blitterswijk CA, Ponec M. Effect of fibroblasts on epidermal regeneration. Br J Dermatol 2002; 147:230-43. [PMID: 12174092 DOI: 10.1046/j.1365-2133.2002.04871.x] [Citation(s) in RCA: 205] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND There is little information on specific interactions between dermal fibroblasts and epidermal keratinocytes. The use of engineered skin equivalents consisting of organotypic cocultures of keratinocytes and fibroblasts offers an attractive approach for such studies. OBJECTIVES To examine the role fibroblasts play in generation and maintenance of reconstructed epidermis. METHODS Human keratinocytes were seeded on collagen matrices populated with increasing numbers of fibroblasts and cultured for 2 weeks at the air-liquid interface. RESULTS In the absence of fibroblasts, stratified epidermis with only three or four viable cell layers was formed. In the presence of fibroblasts, keratinocyte proliferation was stimulated and epidermal morphology was improved. Epidermal morphogenesis was also markedly improved in epidermis generated in organotypic keratinocyte monocultures grown in medium derived from dermal equivalents or from organotypic keratinocyte-fibroblast cocultures. These observations clearly indicate the proliferation-stimulating activity of soluble factors released from fibroblasts. Under all experimental conditions, onset of keratinocyte differentiation was shown by the expression of keratin 10 in all suprabasal cell layers. With increasing numbers of fibroblasts incorporated into the collagen matrix, the expression of markers associated with keratinocyte activation, e.g. keratins 6, 16 and 17 and the cornified envelope precursor SKALP decreased, and involucrin localization shifted toward the granulosum layer. This fibroblast-mediated effect was even more pronounced when the fibroblasts were precultured in the collagen matrices for 1 week instead of overnight. The basement membrane proteins collagen VII and laminin 5 were present at the epithelial-matrix border. The expression of integrin alpha 6 beta 4 and of E-cadherin was comparable with that seen in native skin and was not significantly modulated by fibroblasts. Under all experimental conditions the expression of integrin subunits alpha 2, alpha 3 and beta 1 was upregulated, indicating keratinocyte activation. CONCLUSIONS Our results illustrate that numbers of fibroblasts in the collagen matrix and their functional state is a critical factor for establishment of normal epidermal morphogenesis.
Collapse
Affiliation(s)
- A el-Ghalbzouri
- Department of Dermatology, Leiden University Medical Centre, Sylvius Laboratory, PO Box 9503, 2300 RA Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
182
|
Nowinski D, Höijer P, Engstrand T, Rubin K, Gerdin B, Ivarsson M. Keratinocytes inhibit expression of connective tissue growth factor in fibroblasts in vitro by an interleukin-1alpha-dependent mechanism. J Invest Dermatol 2002; 119:449-55. [PMID: 12190869 DOI: 10.1046/j.1523-1747.2002.01841.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The wound healing process concludes with downregulation of fibroblast activity. Clinical observations suggest that the regenerating epidermis suppresses this activity. An important regulator of fibroblast activity is the fibrogenic cytokine connective tissue growth factor. We hypothesized that epidermal keratinocytes may affect fibroblast activity via this cytokine. We demonstrate keratinocyte-mediated suppression of connective tissue growth factor at both the mRNA and protein levels by around 50% or more when fibroblasts were cultured in multiwell plates with keratinocyte cultures in accompanying semipermeable cell culture inserts, or stimulated by keratinocyte-conditioned media. Both basal and transforming-growth-factor-beta1-stimulated levels of connective tissue growth factor were inhibited. A 3 h coculture period with keratinocytes was sufficient to suppress connective tissue growth factor expression by fibroblasts, but the inhibition developed over a time period of around 16 h. The putative keratinocyte-derived factor(s) responsible for these effects was found to be soluble and stable. By analyzing cytokines secreted by keratinocytes we identified interleukin-1alpha as a potent inhibitor of connective tissue growth factor mRNA expression in fibroblasts. Involvement of this cytokine in keratinocyte-mediated connective tissue growth factor suppression was confirmed by using anti-interleukin-1alpha antibodies. Tumor necrosis factor alpha or prostaglandins did not appear to be involved. In conclusion, our results indicate that interleukin-1alpha secretion by keratinocytes provides a mechanism for the downregulation of connective tissue activity during the end-stage of wound healing, when epithelia coverage has developed over the wound area.
Collapse
Affiliation(s)
- Daniel Nowinski
- Department of Surgical Sciences, Plastic Surgery Unit, and Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | | | | | | | | | | |
Collapse
|
183
|
Lim IJ, Phan TT, Bay BH, Qi R, Huynh H, Tan WTL, Lee ST, Longaker MT. Fibroblasts cocultured with keloid keratinocytes: normal fibroblasts secrete collagen in a keloidlike manner. Am J Physiol Cell Physiol 2002; 283:C212-22. [PMID: 12055090 DOI: 10.1152/ajpcell.00555.2001] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Keloid scars represent a pathological response to cutaneous injury, reflecting a new set point between synthesis and degradation biased toward extracellular matrix (ECM) collagen accumulation. Using a serum-free two-chamber coculture model, we recently demonstrated a significant increase in normal fibroblast proliferation when cocultured with keloid-derived keratinocytes. We hypothesized that similar keratinocyte-fibroblast interactions might influence fibroblast collagen production and examined conditioned media and cell lysate from coculture for collagen I and III production by Western blot, allied with Northern analysis for procollagen I and III mRNA. Normal fibroblasts cocultured with keloid keratinocytes produced increased soluble collagen I and III with a corresponding increase in procollagen I and III mRNA transcript levels. This was associated with decreased insoluble collagen from cell lysate. When keloid fibroblasts were cocultured with keloid keratinocytes, both soluble and insoluble collagen were increased with associated procollagen III mRNA upregulation. Transmission electron microscopy of normal fibroblasts cocultured with keloid keratinocytes showed an ECM appearance similar to in vivo keloid tissue, an appearance not seen when normal fibroblasts were cocultured with normal keratinocytes.
Collapse
Affiliation(s)
- Ivor Jiun Lim
- Division of Plastic Surgery, Department of Surgery, National University of Singapore, Singapore 119074.
| | | | | | | | | | | | | | | |
Collapse
|
184
|
Flanders KC, Sullivan CD, Fujii M, Sowers A, Anzano MA, Arabshahi A, Major C, Deng C, Russo A, Mitchell JB, Roberts AB. Mice lacking Smad3 are protected against cutaneous injury induced by ionizing radiation. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:1057-68. [PMID: 11891202 PMCID: PMC1867194 DOI: 10.1016/s0002-9440(10)64926-7] [Citation(s) in RCA: 235] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/13/2001] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-beta (TGF-beta) plays a central role in the pathogenesis of inflammatory and fibrotic diseases, including radiation-induced fibrosis. We previously reported that mice null for Smad3, a key downstream mediator of TGF-beta, show accelerated healing of cutaneous incisional wounds with reduced inflammation and accumulation of matrix. To determine if loss of Smad3 decreases radiation-induced injury, skin of Smad3+/+ [wild-type (WT)] and -/- [knockout (KO)] mice was exposed to a single dose of 30 to 50 Gy of gamma-irradiation. Six weeks later, skin from KO mice showed significantly less epidermal acanthosis and dermal influx of mast cells, macrophages, and neutrophils than skin from WT littermates. Skin from irradiated KO mice exhibited less immunoreactive TGF-beta and fewer myofibroblasts, suggesting that these mice will have a significantly reduced fibrotic response. Although irradiation induced no change in the immunohistochemical expression of the TGF-beta type I receptor, the epidermal expression of the type II receptor was lost after irradiation whereas its dermal expression remained high. Primary keratinocytes and dermal fibroblasts prepared from WT and KO mice showed similar survival when irradiated, as did mice exposed to whole-body irradiation. These results suggest that inhibition of Smad3 might decrease tissue damage and reduce fibrosis after exposure to ionizing irradiation.
Collapse
Affiliation(s)
- Kathleen C Flanders
- Laboratory of Cell Regulation and Carcinogenesis and Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-5055, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Artuc M, Steckelings UM, Henz BM. Mast cell-fibroblast interactions: human mast cells as source and inducers of fibroblast and epithelial growth factors. J Invest Dermatol 2002; 118:391-5. [PMID: 11874475 DOI: 10.1046/j.0022-202x.2001.01705.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
As mast cells have been implicated in cutaneous repair processes, we have examined the ability of human mast cells to produce important epithelial and fibroblast growth factors or to stimulate the production of such factors in dermal fibroblasts. Isolated, highly purified human dermal mast cells and human leukemic mast cells were examined for mRNA and partly also for protein expression of these molecules as such or after preincubation with interleukin-4, stem cell factor, or with phorbol myristate acetate. In addition, mast cells were studied for their ability to induce fibroblast growth factor 2 and fibroblast growth factor 7 secretion from dermal fibroblasts. Both dermal and leukemic mast cells expressed fibroblast growth factor 2, fibroblast growth factor 7, and heparin-binding epidermal growth factor, but not hepatocyte growth factor at mRNA level, and dermal mast cells expressed fibroblast growth factor 10 in addition. At protein level, spontaneous fibroblast growth factor 2 secretion was noted that was markedly enhanced by phorbol myristate acetate, whereas no fibroblast growth factor 7 protein was detected under these conditions. Instead, human mast cell-1 supernatants induced enhanced fibroblast growth factor 7 secretion from dermal fibroblasts, with phorbol-myristate-acetate-stimulated supernatants being more effective. This effect could be reproduced with histamine and was H1-receptor mediated. Tryptase was ineffective but stimulated instead fibroblast growth factor 2 secretion from fibroblasts. These data demonstrate for the first time the ability of mast cells to express and/or secrete several growth factors of the fibroblast growth factor family as well as heparin-binding epidermal growth factor directly or indirectly via stimulation of fibroblasts, underlining the potentially pivotal role of these cells during human tissue repair and homeostasis.
Collapse
Affiliation(s)
- Metin Artuc
- Department of Dermatology and Allergy, Humboldt University, Charité, Berlin, Germany.
| | | | | |
Collapse
|
186
|
Kameda T, Hatakeyama S, Terada K, Sugiyama T. Acceleration of the formation of cultured epithelium using the sonic hedgehog expressing feeder cells. TISSUE ENGINEERING 2001; 7:545-55. [PMID: 11694189 DOI: 10.1089/107632701753213174] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Sonic hedgehog (Shh) regulates the principal possesses in many developmental stages, including the epithelial-mesenchymal interaction. The extraordinary acceleration of signaling by Shh is responsible for the development of human basal cell carcinomas and trichoepitheliomas; they might originate from the very immature keratinocytes, including the stem cells. We tried to utilize the mitogenic effect of Shh to accelerate the formation of cultured epithelium, which is already used in the medical field practically. To this end, we transfected shh cDNA into a Swiss-3T3 cell line, widely used as a feeder for keratinocytes, and established a Shh expressing cell line. The lethally irradiated Shh expressing feeder cells remarkably accelerated the growth of keratinocyte colonies obtained from the human neonatal foreskin, and the formation of well-stratified cultured epithelium, which is rich in immature small keratinocytes, expressing cytokeratin 14. This acceleration was suppressed by the addition of cyclopamine, a specific inhibitor of Shh signaling. These data indicate that the Shh is a promising mitogen to improve the technology for cultured epithelium formation.
Collapse
Affiliation(s)
- T Kameda
- Department of Biochemistry, Akita University School of Medicine, Akita, Japan.
| | | | | | | |
Collapse
|
187
|
Loertscher JA, Sattler CA, Allen-Hoffmann BL. 2,3,7,8-Tetrachlorodibenzo-p-dioxin alters the differentiation pattern of human keratinocytes in organotypic culture. Toxicol Appl Pharmacol 2001; 175:121-9. [PMID: 11543644 DOI: 10.1006/taap.2001.9202] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human exposure to the environmental toxin 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) produces a severe skin pathology known as chloracne. In these studies we employed a three-dimensional, organotypic model system to study the effects of TCDD on human skin. This model uses the spontaneously immortalized human keratinocyte cell line NIKS and recapitulates both the three-dimensional microenvironment and epithelial-mesenchymal interactions found in intact human skin. Treatment of the organotypic cultures with TCDD causes alterations in the pattern of keratinocyte terminal differentiation. Analysis at both the light and electron microscope levels reveals a fully differentiated cornified layer in TCDD-treated organotypic cultures at earlier time points than observed in vehicle (dimethyl sulfoxide)-treated controls. Furthermore, TCDD-treated organotypic cultures exhibit aberrant distribution of several differentiation-specific protein markers. Basal cells in TCDD- and DMSO-treated organotypic cultures show no differences in proliferation as measured by quantification of 5-bromo-2'-deoxyuridine (BrdU)-positive nuclei. No aberrant BrdU uptake was detected outside of the basal layer. Neither TUNEL labeling nor immunohistochemical staining with an antibody to active caspase-3 revealed increased apoptosis in TCDD-treated organotypic cultures relative to controls. These data clearly indicate that TCDD modulates homeostasis in a model of human stratifying epithelium independent of changes in proliferation and apoptosis, exclusively by impacting keratinocyte terminal differentiation. This TCDD-induced effect on differentiation-specific proteins results in profound changes in the tissue architecture.
Collapse
Affiliation(s)
- J A Loertscher
- Department of Pathology, University of Wisconsin Medical School, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
188
|
Maas-Szabowski N, Szabowski A, Stark HJ, Andrecht S, Kolbus A, Schorpp-Kistner M, Angel P, Fusenig NE. Organotypic Cocultures with Genetically Modified Mouse Fibroblasts as a Tool to Dissect Molecular Mechanisms Regulating Keratinocyte Growth and Differentiation. J Invest Dermatol 2001; 116:816-20. [PMID: 11348477 DOI: 10.1046/j.1523-1747.2001.01349.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Organotypic cocultures of keratinocytes and fibroblasts generate a normal epidermis irrespective of the species and tissue origin of fibroblasts. The use of mouse fibroblasts and human keratinocytes facilitates the identification of the origin of compounds involved in epidermal tissue reconstitution and growth regulation. Moreover, the functional significance for the keratinocyte phenotype of genetically modified fibroblasts from transgenic or knockout mice, even those exhibiting an embryonic lethal phenotype, can be studied in such heterologous in vitro tissue equivalents. Here we communicate results of such studies revealing the antagonistic function of mouse fibroblasts defective in the AP-1 constituents c-Jun and JunB, respectively, on human keratinocyte growth and differentiation. Furthermore, the hematopoietic growth factor granulocyte macrophage-colony stimulating factor has been identified as a novel regulator of keratinocyte growth and differentiation. As will be reported in detail elsewhere both granulocyte macrophage-colony stimulating factor and keratinocyte growth factor have been identified as major mediators of fibroblast-keratinocyte interactions and their expression is induced via AP-1 by interleukin-1 released by the epithelial cells. Thus, these heterologous cocultures provide a novel promising tool for elucidating molecular mechanisms of epithelial-mesenchymal interactions and their consequences on epithelial cell proliferation and differentiation.
Collapse
Affiliation(s)
- N Maas-Szabowski
- Division of Carcinogenesis and Differentiation, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
189
|
Abstract
The histoarchitecture and function of the epidermis depend on a well-controlled balance between keratinocyte proliferation and differentiation. This balance is perturbed after skin injury, and imbalance is a characteristic feature of major human skin diseases such as psoriasis and epidermal cancers. Recent studies have highlighted the importance of fibroblast-derived soluble factors for the regulation of keratinocyte proliferation and differentiation. Therefore, identification of these paracrine-acting factors and the elucidation of their mechanisms of action are necessary for understanding epidermal homeostasis, repair and disease, and these approaches will offer new potential targets for drug therapy. Here, we review exciting recent findings on the identification, regulation and function of paracrine-acting cytokines in the skin. In particular, we describe the role of fibroblast-derived mitogens as regulators of keratinocyte proliferation and differentiation, and we summarize the regulation of these factors by keratinocyte-derived interleukin 1 that involves the transcription factors c-Jun and JunB.
Collapse
Affiliation(s)
- S Werner
- Institute of Cell Biology, Swiss Federal Institute of Technology, ETH-Hönggerberg, CH-8093 Zürich, Switzerland.
| | | |
Collapse
|
190
|
Mackenzie IC, Gao Z. Keratinocyte growth factor expression in human gingival fibroblasts and stimulation of in vitro gene expression by retinoic acid. J Periodontol 2001; 72:445-53. [PMID: 11338296 DOI: 10.1902/jop.2001.72.4.445] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Keratinocyte growth factor (KGF) is a stromally derived growth factor of the fibroblast growth factor (FGF) family with paracrine effects targeted to influence the growth and differentiation of epithelia. Regional and temporal changes in KGF expression play important roles in the development and maintenance of epithelial structures and in epithelial wound healing. Differing patterns of expression of KGF by fibroblasts in the gingival region could therefore be related to the observed regional variation in the differentiation and behavior of gingival epithelia. METHODS The in vitro and in vivo patterns of expression of KGF mRNA in human gingival and periodontal fibroblasts were examined using reverse transcription polymerase chain reactions (RT-PCR) and in situ hybridization with digoxigenin-labeled riboprobes. The patterns observed for human gingiva were compared with those for human skin and for murine tissues. RESULTS Gingival and periodontal fibroblasts showed expression of KGF transcripts in vitro, and the degree of expression was markedly influenced by the presence of retinoic acid, an agent known to influence patterns of epithelial differentiation. Sections of human and murine gingiva and skin showed regionally variable expression of transcripts with the cells expressing KGF in the subepithelial, rather than the deeper, connective tissues and periodontium. CONCLUSIONS The results point to a role of KGF in the maintenance of normal growth and differentiation of gingival epithelia. A lack of KGF expression by periodontal fibroblasts in vivo is expected to hinder apical epithelial migration and thus stabilize the epithelial attachment. The effects of retinoic acid (RA) on KGF expression in vitro provide an indirect mechanism by which RA may regulate the growth and differentiation of gingival epithelia.
Collapse
Affiliation(s)
- I C Mackenzie
- Department of Adult Dental Health, University of Wales College of Medicine, Heath Park, Cardiff, UK.
| | | |
Collapse
|
191
|
Bagutti C, Hutter C, Chiquet-Ehrismann R, Fässler R, Watt FM. Dermal fibroblast-derived growth factors restore the ability of beta(1) integrin-deficient embryonal stem cells to differentiate into keratinocytes. Dev Biol 2001; 231:321-33. [PMID: 11237462 DOI: 10.1006/dbio.2000.0149] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Embryonal stem (ES) cells that are homozygous null for the beta(1) integrin subunit fail to differentiate into keratinocytes in vitro but do differentiate in teratomas and wild-type/beta(1)-null chimeric mice. The failure of beta(1)-null ES cells to differentiate in culture might be the result of defective extracellular matrix assembly or reduced sensitivity to soluble inducing factors. By culturing embryoid bodies on dead, deepidermized human dermis (DED) we showed that epidermal basement membrane did not induce beta(1)-null ES cells to undergo keratinocyte differentiation and did not stimulate the differentiation of wild-type ES cells. Coculture with epidermal keratinocytes also had no effect. However, when human dermal fibroblasts were incorporated into DED, the number of epidermal cysts formed by wild-type ES cells increased dramatically, and small groups of keratin 14-positive cells differentiated from beta(1)-null ES cells. Fibroblast-conditioned medium stimulated differentiation of K14-positive cells in wild-type and beta(1)-null embryoid bodies. Of a range of growth factors tested, KGF, FGF10, and TGFalpha all stimulated differentiation of keratin 14-positive beta(1)-null cells, and KGF and FGF10 were shown to be produced by the fibroblasts used in coculture experiments. The effects of the growth factors on wild-type ES cells were much less pronounced, suggesting that the concentrations of inducing factors already present in the medium were not limiting for wild-type cells. We conclude that the lack of beta(1) integrins decreases the sensitivity of ES cells to soluble factors that induce keratinocyte differentiation.
Collapse
Affiliation(s)
- C Bagutti
- Keratinocyte Laboratory, Imperial Cancer Research Fund, 44 Lincoln's Inn Fields, London, WC2A 3PX, United Kingdom
| | | | | | | | | |
Collapse
|
192
|
Lim IJ, Phan TT, Song C, Tan WT, Longaker MT. Investigation of the influence of keloid-derived keratinocytes on fibroblast growth and proliferation in vitro. Plast Reconstr Surg 2001; 107:797-808. [PMID: 11304607 DOI: 10.1097/00006534-200103000-00022] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Keloids are disfiguring, proliferative scars that represent a pathological response to cutaneous injury. The overabundant extracellular matrix formation, largely from collagen deposition, is characteristic of these lesions and has led to investigations into the role of the fibroblast in its pathogenesis. Curiously, the role of the epidermis in extracellular matrix collagen deposition of normal skin has been established, but a similar hypothesis in keloids has not been investigated. The aim of this study was to investigate the influence of keloid epithelial keratinocytes on the growth and proliferation of normal fibroblasts in an in vitro serum-free co-culture system. A permeable membrane separated two chambers; the upper chamber contained a fully differentiated stratified epithelium derived from the skin of excised earlobe keloid specimens, whereas the lower chamber contained a monolayer of normal or keloid fibroblasts. Both cell types were nourished by serum-free medium from the lower chamber. Epithelial keratinocytes from five separate earlobe keloid specimens were investigated. Four sets of quadruplicates were performed for each specimen co-cultured with normal fibroblasts or keloid-derived fibroblasts. Controls consisted of (1) normal keratinocytes co-cultured with normal fibroblasts, and (2) fibroblasts grown in serum-free media in the absence of keratinocytes in the upper chamber. Fibroblasts were indirectly quantified by 3- (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide colorimetric assay, with results confirmed by DNA content measurement, at days 1 and 5 after the co- culture initiation.Significantly, increased proliferation was seen in fibroblasts co-cultured with keloid keratinocytes, as compared with the normal keratinocyte controls at day 5 (analysis of variance, p < 0.001). These results strongly suggest that the overlying epidermal keratinocytes of the keloid may have an important, previously unappreciated role in keloid pathogenesis using paracrine or epithelial-mesenchymal signaling.
Collapse
Affiliation(s)
- I J Lim
- Division of Plastic Surgery, the Department of Surgery, National University Hospital, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
193
|
Johnen C, Kage A, Oechel A, Neuhaus P, Gerlach JC. Investigation on reconstruction of skin for autologous transplantation. Transplant Proc 2001; 33:618-9. [PMID: 11266985 DOI: 10.1016/s0041-1345(00)02169-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- C Johnen
- Department of Surgery, Berlin, Germany
| | | | | | | | | |
Collapse
|
194
|
Hoffmann J, Twiesselmann C, Kummer MP, Romagnoli P, Herzog V. A possible role for the Alzheimer amyloid precursor protein in the regulation of epidermal basal cell proliferation. Eur J Cell Biol 2000; 79:905-14. [PMID: 11152291 DOI: 10.1078/0171-9335-00117] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The regulation of epidermal growth involves a number of ions, growth factors and cytokines and possibly additional but as yet unknown factors. Here we report on the potential role of the secretory N-terminal domain (sAPP) of the Alzheimer amyloid precursor protein (APP) in the regulation of keratinocyte proliferation. In human skin APP was detectable predominantly in the basal cell layer of the epidermis whereas the immunocytochemical signal in the underlying mesenchymal tissue was very low. Cultured normal human keratinocytes expressed the three APP isoforms 695, 751 and 770 with highest values for the isoforms 751 and 770. HaCaT cells, a spontaneously immortalized human keratinocyte cell line, exhibited almost identical patterns in the expression of the APP isoforms and in the release of endogenous sAPP. In HaCaT cells, recombinant sAPP (sAPPrec) was found to compete with endogenous sAPP for the same binding sites. Binding of sAPPrec was specific and occurred in microdomains of approximately 0.1 to approximately 0.3 microm in diameter. At 10 nM, sAPPrec binding induced a 2- to 4-fold increase in the rate of cell growth. sAPP concentrations in the conditioned media were found to reach 5-20 nM which is in the mitogenic range of sAPPrec. The proliferative effect of sAPP was inhibited by approximately 50% when antisense oligonucleotides directed against the APP mRNA were applied. The predominant expression of
Collapse
Affiliation(s)
- J Hoffmann
- Institut für Zellbiologie und Bonner Forum Biomedizin, Universität Bonn, Germany
| | | | | | | | | |
Collapse
|
195
|
Szabowski A, Maas-Szabowski N, Andrecht S, Kolbus A, Schorpp-Kistner M, Fusenig NE, Angel P. c-Jun and JunB antagonistically control cytokine-regulated mesenchymal-epidermal interaction in skin. Cell 2000; 103:745-55. [PMID: 11114331 DOI: 10.1016/s0092-8674(00)00178-1] [Citation(s) in RCA: 341] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Interactions between mesenchymal and epithelial cells are responsible for organogenesis and tissue homeostasis. This mutual cross-talk involves cell surface proteins and soluble factors, which are mostly the result of regulated transcription. To elucidate dimer-specific functions of the AP-1 family of transcription factors, we reconstituted skin by combining primary human keratinocytes and mouse wild-type, c-jun(-/-), and junB(-/-) fibroblasts. We have discovered an antagonistic function of these AP-1 subunits in the fibroblast-mediated paracrine control of keratinocyte proliferation and differentiation, and traced this effect to the IL-1-dependent regulation of KGF and GM-CSF. These data suggest that the relative activation state of these AP-1 subunits in a non-cell-autonomous, transregulatory fashion directs regeneration of the epidermis and maintenance of tissue homeostasis in skin.
Collapse
Affiliation(s)
- A Szabowski
- Division of Signal Transduction and Growth Control Deutsches Krebsforschungszentrum Im Neuenheimer Feld 280 69120, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
196
|
Maas-Szabowski N, Stark HJ, Fusenig NE. Keratinocyte growth regulation in defined organotypic cultures through IL-1-induced keratinocyte growth factor expression in resting fibroblasts. J Invest Dermatol 2000; 114:1075-84. [PMID: 10844548 DOI: 10.1046/j.1523-1747.2000.00987.x] [Citation(s) in RCA: 165] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Balanced keratinocyte proliferation and differentiation resulting in regular tissue organization strictly depend on dermal support. Organotypic cultures represent biologically relevant in vitro models to study the molecular mechanism of the underlying dermal-epidermal interactions. To mimic the state of resting fibroblasts in the dermis, postmitotic (irradiated) fibroblasts were incorporated in the collagen matrix, where they typically support epidermal proliferation and tissue organization. In coculture with keratinocytes, fibroblasts exhibit an enhanced expression of keratinocyte growth factor and the interleukin-1 receptor (type I), which further increase with culture time. In cocultured keratinocytes, keratinocyte growth factor receptor as well as RNA expression and protein release of interleukin-1alpha and interleukin-1beta are upregulated. We hypothesized that the modulated cytokine expression represents a basic mechanism for keratinocyte growth regulation. The functional significance of this double paracrine pathway, i.e., induction of keratinocyte growth factor expression in fibroblasts by keratinocytes via release of interleukin-1, was confirmed by interfering with both signaling elements: (i) interleukin-1-neutralizing antibodies and interleukin-1 receptor antagonist significantly inhibited keratinocyte growth factor release, keratinocyte proliferation, and tissue formation comparable to the effect produced by keratinocyte-growth-factor-blocking antibodies; (ii) addition of keratinocyte growth factor to cocultures with inactivated interleukin-1 pathway completely reverted growth inhibition; (iii) in organotypic cocultures with subthreshold fibroblast numbers both interleukin-1 and keratinocyte growth factor restored the impaired epidermal morphogenesis. Thus, epidermal tissue regeneration in organotypic cocultures is mainly regulated by keratinocyte-derived interleukin-1 signaling, which induces keratinocyte growth factor expression in cocultured fibroblasts. This demonstrates a novel role for interleukin-1 in skin homeostasis substantiating data from wound healing studies in vivo.
Collapse
Affiliation(s)
- N Maas-Szabowski
- Division of Differentiation and Carcinogenesis, German Cancer Research Center, Heidelberg, Germany
| | | | | |
Collapse
|