Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cells ; 12(12)2023 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-37371048

RESUMEN

Amniotic fluid has been proposed as an easily available source of cells for numerous applications in regenerative medicine and tissue engineering. The use of amniotic fluid cells in biomedical applications necessitates their unequivocal characterization; however, the exact cellular composition of amniotic fluid and the precise tissue origins of these cells remain largely unclear. Using cells cultured from the human amniotic fluid of fetuses with spina bifida aperta and of a healthy fetus, we performed single-cell RNA sequencing to characterize the tissue origin and marker expression of cultured amniotic fluid cells at the single-cell level. Our analysis revealed nine different cell types of stromal, epithelial and immune cell phenotypes, and from various fetal tissue origins, demonstrating the heterogeneity of the cultured amniotic fluid cell population at a single-cell resolution. It also identified cell types of neural origin in amniotic fluid from fetuses with spina bifida aperta. Our data provide a comprehensive list of markers for the characterization of the various progenitor and terminally differentiated cell types in cultured amniotic fluid. This study highlights the relevance of single-cell analysis approaches for the characterization of amniotic fluid cells in order to harness their full potential in biomedical research and clinical applications.


Asunto(s)
Espina Bífida Quística , Disrafia Espinal , Humanos , Líquido Amniótico/metabolismo , Espina Bífida Quística/metabolismo , Análisis de Expresión Génica de una Sola Célula , Disrafia Espinal/metabolismo , Ingeniería de Tejidos
2.
Cells ; 11(14)2022 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-35883599

RESUMEN

The basal layer of human interfollicular epidermis has been described to harbour both quiescent keratinocyte stem cells and a transit amplifying cell population that maintains the suprabasal epidermal layers. We performed immunofluorescence analyses and revealed that the main proliferative keratinocyte pool in vivo resides suprabasally. We isolated from the human epidermis two distinct cell populations, the basal and the suprabasal keratinocytes, according to the expression of integrin ß4 (iß4). We compared basal iß4+ or suprabasal iß4- keratinocytes with respect to their proliferation and colony-forming ability and their Raman spectral properties. In addition, we generated dermo-epidermal substitutes using freshly isolated and sorted basal iß4+ or suprabasal iß4- keratinocytes and transplanted them on immuno-compromised rats. We show that suprabasal iß4- keratinocytes acquire a similar proliferative capacity as basal iß4+ keratinocytes after two weeks of culture in vitro, with expression of high levels of iß4 and downregulation of K10 expression. In addition, both basal iß4+ and suprabasal iß4- keratinocytes acquire authentic self-renewing properties during the in vitro 3D-culture phase and are able to generate and maintain a fully stratified epidermis for 16 weeks in vivo. Therefore, against the leading dogma, we propose that human suprabasal keratinocytes can retro-differentiate into true basal stem cells in a wound situation and/or when in contact with the basement membrane.


Asunto(s)
Piel Artificial , Animales , Dermis , Células Epidérmicas , Epidermis/metabolismo , Humanos , Queratinocitos/metabolismo , Ratas
3.
J Tissue Eng ; 13: 20417314221088513, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35495096

RESUMEN

Extensive availability of engineered autologous dermo-epidermal skin substitutes (DESS) with functional and structural properties of normal human skin represents a goal for the treatment of large skin defects such as severe burns. Recently, a clinical phase I trial with this type of DESS was successfully completed, which included patients own keratinocytes and fibroblasts. Yet, two important features of natural skin were missing: pigmentation and vascularization. The first has important physiological and psychological implications for the patient, the second impacts survival and quality of the graft. Additionally, accurate reproduction of large amounts of patient's skin in an automated way is essential for upscaling DESS production. Therefore, in the present study, we implemented a new robotic unit (called SkinFactory) for 3D bioprinting of pigmented and pre-vascularized DESS using normal human skin derived fibroblasts, blood- and lymphatic endothelial cells, keratinocytes, and melanocytes. We show the feasibility of our approach by demonstrating the viability of all the cells after printing in vitro, the integrity of the reconstituted capillary network in vivo after transplantation to immunodeficient rats and the anastomosis to the vascular plexus of the host. Our work has to be considered as a proof of concept in view of the implementation of an extended platform, which fully automatize the process of skin substitution: this would be a considerable improvement of the treatment of burn victims and patients with severe skin lesions based on patients own skin derived cells.

4.
Sci Rep ; 12(1): 1944, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35121765

RESUMEN

CD26, also known as dipeptidyl peptidase IV (DPPIV), is a multifunctional transmembrane protein playing a significant role in the cutaneous wound healing processes in the mouse skin. However, only scarce data are available regarding the distribution and function of this protein in the human skin. Therefore, the aim of this study was to investigate the impact of CD26 deficiency in human primary fibroblasts on the regeneration of human tissue-engineered skin substitutes in vivo. Dermo-epidermal skin analogs, based on collagen type I hydrogels, were populated either with human CD26+ or CD26knockout fibroblasts and seeded with human epidermal keratinocytes. These skin substitutes were transplanted onto the back of immune-incompetent rodents. Three weeks post-transplantation, the grafts were excised and analyzed with respect to specific epidermal and dermal maturation markers. For the first time, we show here that the expression of CD26 protein in human dermis is age-dependent. Furthermore, we prove that CD26+ fibroblasts are more active in the production of extracellular matrix (ECM) both in vitro and in vivo and are necessary to achieve rapid epidermal and dermal homeostasis after transplantation.


Asunto(s)
Comunicación Celular , Proliferación Celular , Dipeptidil Peptidasa 4/metabolismo , Fibroblastos/trasplante , Queratinocitos/trasplante , Regeneración , Trasplante de Piel , Piel Artificial , Adolescente , Animales , Células Cultivadas , Niño , Preescolar , Técnicas de Cocultivo , Dipeptidil Peptidasa 4/deficiencia , Dipeptidil Peptidasa 4/genética , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/enzimología , Xenoinjertos , Humanos , Lactante , Queratinocitos/metabolismo , Masculino , Ratas Desnudas , Transducción de Señal , Factores de Tiempo
5.
Biomaterials ; 273: 120779, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33932701

RESUMEN

The clinical treatment of large, full-thickness skin injuries with tissue-engineered autologous dermo-epidermal skin substitutes is an emerging alternative to split-thickness skin grafting. However, their production requires about one month of in vitro cell and tissue culture, which is a significant drawback for the treatment of patients with severe skin defects. With the aim to reduce the production time, we developed a new dynamic bioreactor setup that applies cyclic biaxial tension to collagen hydrogels for skin tissue engineering. By reliably controlling the time history of mechanical loading, the dynamic culturing results in a three-fold increase in collagen hydrogel stiffness and stimulates the embedded fibroblasts to enter the cell cycle. As a result, the number of fibroblasts is increased by 75% compared to under corresponding static culturing. Enhanced fibroblast proliferation promotes expression of dermal extracellular matrix proteins, keratinocyte proliferation, and the early establishment of the epidermis. The time required for early tissue maturation can therefore be reduced by one week. Analysis of the separate effects of cyclic loading, matrix stiffening, and interstitial fluid flow indicates that cyclic deformation is the dominant biophysical factor determining fibroblast proliferation, while tissue stiffening plays a lesser role. Local differences in the direction of deformation (in-plane equibiaxial vs. uniaxial strain) influence fibroblast orientation but not proliferation, nor the resulting tissue properties. Importantly, dynamic culturing does not activate fibroblast differentiation into myofibroblasts. The present work demonstrates that control of mechanobiological cues can be very effective in driving cell response toward a shorter production time for human skin substitutes.


Asunto(s)
Piel Artificial , Proliferación Celular , Dermis , Fibroblastos , Humanos , Piel , Trasplante de Piel , Ingeniería de Tejidos
6.
J Biomed Mater Res B Appl Biomater ; 108(7): 3045-3055, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32420687

RESUMEN

OBJECTIVE: The gold standard for bladder regeneration in end-stage bladder disease is the use of intestinal tissue, which is however associated with significant long-term complications. Our study aims to bioengineer functional detrusor muscle combining bladder smooth muscle cells (SMC) and SMC-like adipose-derived stem cells (pADSC) in compressed collagen (CC) hydrogels and to investigate biocompatibility and tissue regeneration of such detrusor-equivalents in a rat detrusorectomy model. METHODS: Compressed collagen hydrogels seeded with 1 × 106 or 4 × 106 SMC alone or in combination with pADSC in a 1:1 ratio were investigated. Morphology, phenotype, and viability as well as proteomic secretome analysis were assessed in the 1:1 co-cultures and the respective monocultures. The hydrogels were implanted into rat bladders after partial detrusorectomy. Bladders were harvested 8 weeks after transplantation, and assessed for tissue morphology, detrusor regeneration, neo-vascularization and -innervation. RESULTS: Co-cultured cells exhibited native SMC morphology, high viability and proliferated to form microtissues in vitro. The pro-angiogenic factors angiogenin, vascular endothelial growth factor (VEGF)-A and -D were increased in the secretome of the pADSC samples. After 8 weeks of in vivo, the regenerated bladder wall showed a multilayered structure containing all bladder wall components. The overall performance of the bladder wall regeneration of CC seeded with 4 × 106 cells was significantly better than with 1 × 106 cells and the combination SMC:pADCS performed slightly better than SMC alone. CONCLUSION: Compressed collagen possesses an adequate regenerative potential to promote regeneration of bladder wall tissue in vivo. Seeded with a combination of pADSC and SMC this may well be the first step towards a functional bladder reconstruction especially in patients suffering of end-stage bladder diseases.


Asunto(s)
Colágeno , Hidrogeles , Miocitos del Músculo Liso/metabolismo , Regeneración , Ingeniería de Tejidos , Vejiga Urinaria/fisiología , Animales , Células Cultivadas , Femenino , Hidrogeles/química , Hidrogeles/farmacología , Ratas , Ratas Endogámicas Lew , Vejiga Urinaria/lesiones
7.
J Tissue Eng Regen Med ; 14(1): 58-65, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31595702

RESUMEN

An intricate problem during open human fetal surgery for spina bifida regards back skin closure, particularly in those cases where the skin defect is much too large for primary closure. We hypothesize that tissue engineering of fetal skin might provide an adequate autologous skin substitute for in utero application in such situations. Eight sheep fetuses of four time-mated ewes underwent fetoscopic skin biopsy at 65 days of gestation. Fibroblasts and keratinocytes isolated from the biopsy were used to create fetal dermo-epidermal skin substitutes. These were transplanted on the fetuses by open fetal surgery at 90 days of gestation on skin defects (excisional wounds) created during the same procedure. Pregnancy was allowed to continue until euthanasia at 120 days of gestation. The graft area was analyzed macroscopically and microscopically. The transplanted fetal dermo-epidermal skin substitutes was well discernable in situ in three of the four fetuses available for analysis. Histology confirmed healed grafts with a close to natural histological skin architecture four weeks after in utero transplantation. This experimental study generates evidence that laboratory grown autologous fetal skin analogues can successfully be transplanted in utero. These results have clinical implications as an analogous procedure might be applied in human fetuses undergoing prenatal repair to facilitate primary skin closure. Finally, this study may also fertilize the field of fetal tissue engineering in general, particularly when more interventional, minimally invasive, and open fetal surgical procedures become available.


Asunto(s)
Trasplante de Células/métodos , Fetoscopía/métodos , Trasplante de Piel/métodos , Disrafia Espinal/cirugía , Animales , Ingeniería Biomédica , Biopsia , Epidermis , Fibroblastos/citología , Queratinocitos/citología , Riesgo , Ovinos , Silicio/química , Piel/patología , Piel Artificial , Ingeniería de Tejidos/métodos , Andamios del Tejido
8.
Pediatr Surg Int ; 36(1): 25-31, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31576465

RESUMEN

PURPOSE: During in utero surgical spina bifida repair, a multi-layer closure is used to cover the defect. These soft tissues, however, might be not sufficient to protect the spinal cord during the future life. Our goal is to develop a more rigid protective tissue construct consisting of bioengineered cartilage and skin. METHODS: Ovine fetal chondrocytes were tested for their in vitro chondrogenic potential in three-dimensional cultures. Scaffolds based on natural biopolymers (collagen I, fibrin glue) were loaded with varying amounts of fetal chondrocytes and assessed for their ability to support cartilage formation in vitro. The bioengineered constructs were analyzed using cartilage-specific histology stainings and compared to native fetal cartilage. RESULTS: Fetal chondrocytes actively produced cartilage extracellular matrix in three-dimensional cultures, even at high passages. Among all bioengineered scaffolds, only the collagen I-based hydrogels loaded with high densities of fetal chondrocytes showed cartilage-like structure in vitro but also extensive shrinking. CONCLUSION: Fetal chondrocytes represent a good cell source for cartilage bioengineering. Collagen I scaffolds support cartilage formation in vitro, but the construct shrinking constitutes a major limitation. Future steps include the identification of suitable bioprintable materials which maintain their shape and size, as well as the analysis of the interphase between bioengineered cartilage and skin.


Asunto(s)
Cartílago/fisiología , Condrocitos/citología , Feto/citología , Disrafia Espinal/cirugía , Ingeniería de Tejidos/métodos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Feto/cirugía , Humanos , Ovinos , Andamios del Tejido
9.
Fetal Diagn Ther ; 47(3): 198-204, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31509837

RESUMEN

PURPOSE: Open fetal spina bifida repair has become a novel clinical standard of care. In very large spina bifida lesions, the skin defect cannot be covered primarily, asking for alternative solutions. We hypothesize that amniotic fluid stem cells (AFSC) could be differentiated into keratinocytes that could then be used to bioengineer autologous skin usable for in utero back coverage. METHODS: To obtain human AFSC, amniotic fluid samples obtained from fetal surgeries were subjected to immunoselection for c-kit. C-kit-positive samples and controls were cultured with the additives morphogenetic protein 4 and vitamin C to induce differentiation towards keratinocytes. This process was monitored by measuring the expression of K8 and K14 via immunohistochemical staining, flow cytometry, and polymerase chain reaction. RESULTS: After immunoselection and expansion, most cells were positive for K8, but none for K14. After completion of the differentiation protocol, cell colonies with keratinocyte-like appearance could be observed, but cells remained positive for K8 and negative for K14, indicating failed differentiation into keratinocytes. CONCLUSIONS: Culturing of keratinocyte-like cells from AFSC, harvested intraoperatively, was not feasible in this setting. The reasons for failure must be investigated and eliminated, as bioengineering of fetal skin for clinical use during fetal surgery for spina bifida remains an attractive goal.


Asunto(s)
Líquido Amniótico/citología , Diferenciación Celular , Queratinocitos/citología , Ingeniería de Tejidos , Humanos , Piel , Espina Bífida Quística/cirugía
10.
Plast Reconstr Surg ; 144(1): 188-198, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31246829

RESUMEN

BACKGROUND: The management of deep partial-thickness and full-thickness skin defects remains a significant challenge. Particularly with massive defects, the current standard treatment, split-thickness skin grafting, is fraught with donor-site limitations and unsatisfactory long-term outcomes. A novel, autologous, bioengineered skin substitute was developed to address this problem. METHODS: To determine whether this skin substitute could safely provide permanent defect coverage, a phase I clinical trial was performed at the University Children's Hospital Zurich. Ten pediatric patients with acute or elective deep partial- or full-thickness skin defects were included. Skin grafts of 49 cm were bioengineered using autologous keratinocytes and fibroblasts isolated from a patient's small skin biopsy specimen (4 cm), incorporated in a collagen hydrogel. RESULTS: Graft take, epithelialization, infection, adverse events, skin quality, and histology were analyzed. Median graft take at 21 days postoperatively was 78 percent (range, 0 to 100 percent). Healed skin substitutes were stable and skin quality was nearly normal. There were four cases of hematoma leading to partial graft loss. Histology at 3 months revealed a well-stratified epidermis and a dermal compartment comparable to native skin. Mean follow-up duration was 15 months. CONCLUSIONS: In the first clinical application of this novel skin substitute, safe coverage of skin defects was achieved. Safety and efficacy phase II trials comparing the novel skin substitute to split-thickness skin grafts are ongoing. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, IV.


Asunto(s)
Quemaduras/cirugía , Trasplante de Piel/métodos , Piel Artificial , Piel/lesiones , Adolescente , Bioingeniería , Células Cultivadas , Niño , Preescolar , Dermis/citología , Dermis/trasplante , Células Epidérmicas/trasplante , Epidermis/trasplante , Femenino , Fibroblastos/trasplante , Humanos , Lactante , Queratinocitos/trasplante , Masculino , Estudios Prospectivos , Trasplante Autólogo
11.
J Allergy Clin Immunol ; 143(1): 292-304.e8, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29775636

RESUMEN

BACKGROUND: Mutations in tetratricopeptide repeat domain 7A (TTC7A) and its mouse orthologue, Ttc7, result in a multisystemic disease, mostly affecting the epithelial barriers and immune system. Despite successful hematopoietic stem cell transplantation, ongoing progression of gastrointestinal manifestations can be life-threatening in TTC7A-deficient patients. OBJECTIVE: We sought to identify whether TTC7A mutations dysregulate epithelial cells only or whether a cell-intrinsic defect in lymphocytes or other cells contributes to disease manifestations. METHODS: Ttc7-mutated (Ttc7fsn/fsn) mice were crossed to generate double-mutant (Rag2-/-Ttc7fsn/fsn) and triple-mutant (Rag2-/-IL2rg-/-Ttc7fsn/fsn) mice. These models, together with bone marrow chimeras, were used to explore the role of adaptive and innate lymphocytes in the flaky skin phenotype. The effect of the Ttc7fsn/fsn mutation on stromal cells was tested in a xenograft model in conjunction with transcriptomic analysis of Ttc7fsn/fsn fibroblasts. RESULTS: We observed that the severity of epithelial hyperproliferation was accentuated by lymphocytes, whereas the phenotype was not induced by transfer of Ttc7-mutated hematopoietic cells. Furthermore, mice completely lacking the lymphocytic compartment were not protected from epithelial hyperproliferation. Ttc7-mutated mouse fibroblasts expressed increased transcript levels of insulin-like growth factor 1 (Igf1) and the antimicrobial protein regenerating islet-derived protein 3γ (Reg3γ). In a xenograft model Ttc7-mutated fibroblasts markedly increased epithelial proliferation of keratinocytes. Thus Ttc7-mutated fibroblasts were identified as potent instigators of epithelial hyperproliferation. CONCLUSION: Our results reveal a previously unsuspected fundamental cell-extrinsic role of Ttc7. We have identified potential candidates for molecularly targeted treatment strategies that will need to be evaluated in future preclinical studies.


Asunto(s)
Proliferación Celular , Dermatitis/inmunología , Células Epiteliales/inmunología , Fibroblastos/inmunología , Enfermedades Genéticas Congénitas/inmunología , Linfocitos/inmunología , Mutación , Proteínas/inmunología , Animales , Células 3T3 BALB , Dermatitis/genética , Dermatitis/patología , Células Epiteliales/patología , Fibroblastos/patología , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Humanos , Linfocitos/patología , Ratones , Ratones Noqueados , Proteínas/genética
12.
Pediatr Surg Int ; 35(1): 129-136, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30430280

RESUMEN

PURPOSE: Ultraviolet (UV) radiation adversely affects skin health at cellular and molecular levels. Hence, UV radiation can directly induce inflammatory responses in the dermis by inducing erythema, edema, inflammation, dermal fibroblasts alterations, and extracellular matrix modifications. METHODS: Human keratinocytes, melanocytes, and fibroblasts were isolated from skin biopsies, cultured, and expanded in vitro. Fibroblasts were seeded into collagen type I hydrogels that were subsequently covered by keratinocytes and melanocytes. These pigmented dermo-epidermal skin substitutes (pigmDESS) were transplanted for 5 weeks onto full-thickness skin wounds on the back of immuno-incompetent rats, exposed to a single UVB dose of 250 mJ/cm2 or unexposed and excised after 1 week. The effects onto the dermis were assessed regarding cell number, cell phenotype, and cell proliferation. Local inflammation by granulocytes (HIS48) or macrophages (CD11b, iNOS) was analyzed by immunohistochemistry staining. RESULTS: We observed a significantly enhanced ingrowth rate of blood capillaries, but not of lymphatic capillaries at 1 week post-irradiation. Moreover, the enhanced vascularization of pigmDESS after UVB exposure was concomitant with a high infiltration of granulocytes and monocytes/macrophages to the dermal part of grafts. In addition, a heterogeneous expression of HIF-1α and TNFα was detected at this early phase after UVB exposure. In local cellular response examination, results only show a moderate cell proliferation in the dermis. CONCLUSIONS: We were able to define early markers of UVB-induced effects in the dermis of pigmDESS. Overall, a single UVB dose induces temporary acute angiogenic and immune responses during the early post-irradiation phase in vivo.


Asunto(s)
Colágeno Tipo I/metabolismo , Dermis/trasplante , Inflamación/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Trasplante de Piel/métodos , Piel Artificial , Ingeniería de Tejidos/métodos , Animales , Biomarcadores/metabolismo , Biopsia , Células Cultivadas , Dermis/metabolismo , Dermis/efectos de la radiación , Epidermis/metabolismo , Epidermis/efectos de la radiación , Epidermis/trasplante , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Humanos , Inmunohistoquímica , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Ratas , Rayos Ultravioleta
13.
Pediatr Surg Int ; 35(1): 121-127, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30382375

RESUMEN

AIM OF THE STUDY: The use of autologous bio-engineered dermo-epidermal skin substitutes (DESS) yields a pivotal opportunity to cover large skin defects in human patients. These skin grafts consist of both epidermal and dermal compartments necessary for robust and permanent functional wound closure. In this study, we investigated the impact of mesenchymal cells derived from different body site origins on the expression pattern of diverse markers within DESS. METHODS: Human keratinocytes were obtained from interfollicular epidermis, and mesenchymal cells were isolated from foreskin, palmar skin, fat tissue, and tonsils. After expansion, epidermal cells were seeded on collagen I hydrogels containing stromal cells. These human DESS were transplanted on the back of immune-incompetent rats. After 3 weeks, transplants were excised and analyzed using immunohistology techniques. MAIN RESULTS: The macroscopic appearance of skin grafts containing tonsil, fat tissue, or palmar derived mesenchymal cells, was similar to substitutes with foreskin derived dermal fibroblasts. All skin grafts had a strong membrane-localized expression of Lingo-1 in the epidermis. Additionally, we observed an intense expression of transglutaminase 5 in upper epidermal cell layers of the skin grafts confirming a proper keratinocyte differentiation. Tropoelastin was localized throughout the dermal compartments and tightly in contact with the dermo-epidermal junction suggesting an advanced maturation of all skin grafts. CONCLUSIONS: Our data implicate that stromal cells derived from tonsil, fat tissue, and palmar skin can assume fibroblast functions supporting keratinocyte proliferation and differentiation. These findings indicate that distinct types of mesenchymal cells can be clinically used for skin engineering purposes.


Asunto(s)
Dermis/trasplante , Trasplante de Piel/métodos , Piel Artificial , Células del Estroma/citología , Ingeniería de Tejidos/métodos , Adolescente , Adulto , Anciano , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Epidermis/trasplante , Femenino , Fibroblastos/citología , Humanos , Lactante , Queratinocitos/citología , Masculino , Persona de Mediana Edad , Adulto Joven
14.
J Invest Dermatol ; 138(12): 2644-2652, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30096351

RESUMEN

By forming a protective barrier, epidermal keratinocytes represent the first line of defense against environmental insults. UVB radiation of the sun is a major challenge for the skin and can induce inflammation, aging, and eventually skin cancer. UVB induces an immune response in human keratinocytes resulting in activation and secretion of the proinflammatory cytokines proIL-1ß and -18. This is mediated by an assembly of protein complexes, termed inflammasomes. However, the mechanisms underlying sensing of UVB by keratinocytes, and particularly the types of inflammasomes required for cytokine secretion, are a matter of debate. To address these questions, we established a protocol that allows the generation of CRISPR/Cas9-targeted human primary keratinocytes. Our experiments showed an essential role of the NLRP1 rather than the NLRP3 inflammasome in UVB sensing and subsequent IL-1ß and -18 secretion by keratinocytes. Moreover, NLRP1 but not NLRP3 was required for inflammasome activation in response to nigericin, a potassium ionophore and well-established NLRP3 activator in immune cells. Because the CRISPR/Cas9-targeted cells retained their full differentiation capacity, genome editing of human primary keratinocytes might be useful for numerous research and medical applications.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Sistemas CRISPR-Cas , Edición Génica/métodos , Inflamasomas/metabolismo , Queratinocitos/fisiología , Radiodermatitis/genética , Rayos Ultravioleta/efectos adversos , Proteína 9 Asociada a CRISPR/genética , Células Cultivadas , Humanos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas NLR , Nigericina/farmacología , Cultivo Primario de Células , Radiodermatitis/metabolismo
15.
Acta Biomater ; 70: 129-139, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29454158

RESUMEN

Molecular and mechanical interactions with the 3D extracellular matrix are essential for cell functions such as survival, proliferation, migration, and differentiation. Thermo-responsive biomimetic polyisocyanopeptide (PIC) hydrogels are promising new candidates for 3D cell, tissue, and organ cultures. This is a synthetic, thermo-responsive and stress-stiffening material synthesized via polymerization of the corresponding monomers using a nickel perchlorate as a catalyst. It can be tailored to meet various demands of cells by modulating its stiffness and through the decoration of the polymer with short GRGDS peptides using copper free click chemistry. These peptides make the hydrogels biocompatible by mimicking the binding sites of certain integrins. This study focuses on the optimization of the PIC polymer properties for efficient cell, tissue and organ development. Screening for the optimal stiffness of the hydrogel and the ideal concentration of the GRGDS ligand conjugated with the polymer, enabled cell proliferation, migration and differentiation of various primary cell types of human origin. We demonstrate that fibroblasts, endothelial cells, adipose-derived stem cells and melanoma cells, do survive, thrive and differentiate in optimized PIC hydrogels. Importantly, these hydrogels support the spontaneous formation of complex structures like blood capillaries in vitro. Additionally, we utilized the thermo-responsive properties of the hydrogels for a rapid and gentle recovery of viable cells. Finally, we show that organotypic structures of human origin grown in PIC hydrogels can be successfully transplanted subcutaneously onto immune-compromised rats, on which they survive and integrate into the surrounding tissue. STATEMENT OF SIGNIFICANCE: Molecular and mechanical interactions with the surrounding environment are essential for cell functions. Although 2D culture systems greatly contributed to our understanding of complex biological phenomena, they cannot substitute for crucial interaction that take place in 3D. 3D culture systems aim to overcome limitations of the 2D cultures and answer new questions about cell functions. Thermo-responsive biomimetic polyisocyanopeptide (PIC) hydrogels are promising new candidates for 3D cell, tissue, and organ cultures. They are synthetic and can be tailor to meet certain experimental demands. Additionally, they are characterized by strain-stiffening, a feature crucial for cell behaviour, but rare in hydrogels. Their thermos-responsive properties enable quick recovery of the cells by a simple procedure of lowering the temperature.


Asunto(s)
Tejido Adiposo/metabolismo , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Hidrogeles/química , Neovascularización Fisiológica , Péptidos/química , Células Madre/metabolismo , Tejido Adiposo/química , Animales , Línea Celular Tumoral , Química Clic , Células Endoteliales/citología , Femenino , Fibroblastos/citología , Humanos , Ratas , Ratas Desnudas , Células Madre/citología
16.
Cell Death Dis ; 9(2): 24, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29348630

RESUMEN

Inflammasomes are multimeric protein complexes that assemble upon sensing of a variety of stress factors. Their formation results in caspase-1-mediated activation and secretion of the pro-inflammatory cytokines pro-interleukin(IL)-1ß and -18, which induce an inflammatory response. Inflammation is supported by a lytic form of cell death, termed pyroptosis. Innate immune cells, such as macrophages or dendritic cells, express and activate inflammasomes. However, it has also been demonstrated that human primary keratinocytes activate different types of inflammasomes in vitro, for example, upon UVB irradiation or viral infection. Keratinocytes are the main cell type of the epidermis, the outermost layer of the body, and form a protective barrier consisting of a stratified multi-layered epithelium. In human, gain-of-function mutations of the NLRP1 gene cause syndromes mediated by inflammasome activation in keratinocytes that are characterised by skin inflammation and skin cancer susceptibility. Here we demonstrate that murine keratinocytes do not activate inflammasomes in response to stimuli, which induce IL-1ß and -18 secretion by human keratinocytes. Whereas murine keratinocytes produced caspase-1 and proIL-18, expression of the inflammasome proteins Nlrp1, Nlrp3, Aim2, Asc, and proIL-1ß was, compared to human keratinocytes or murine dendritic cells, very low or even undetectable. Priming of murine keratinocytes with cytokines commonly used for induction of proIL-1ß and inflammasome protein expression did not rescue inflammasome activation. Nevertheless, UVB-induced inflammation and neutrophil recruitment in murine skin was dependent on IL-1ß and caspase-1. However, also under these conditions, we did not detect expression of proIL-1ß by keratinocytes in murine skin, but by immune cells. These results demonstrate a higher immunological competence of human compared to murine keratinocytes, which is reflected by stress-induced IL-1ß secretion that is mediated by inflammasomes. Therefore, keratinocytes in human skin can exert immune functions, which are carried out by professional immune cells in murine skin.


Asunto(s)
Inflamasomas/metabolismo , Queratinocitos/metabolismo , Animales , Humanos , Ratones
17.
Nat Commun ; 9(1): 314, 2018 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-29358574

RESUMEN

The originally published version of this Article was updated shortly after publication to add the words 'The' and 'affinity' to the title, following their inadvertent removal during the production process. This has now been corrected in both the PDF and HTML versions of the Article.

18.
J Tissue Eng Regen Med ; 12(2): 505-515, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28715139

RESUMEN

We developed human dermo-epidermal skin substitutes that are presently applied in phase I and II clinical trials. Here, we used these very same skin equivalents containing melanocytes, named MelSkin, as an experimental skin model. We investigated the effects of ultraviolet B (UVB) irradiation on the skin grafts transplanted on immune-compromised rats. The irradiation induces a strong wound healing response going along with massive proliferation of basal keratinocytes, basically quiescent under nonirradiated, homeostatic conditions. As a consequence of UVB irradiation, the initially clearly defined basal keratinocyte (mono)layer expands into about 3 layers of keratinocytes, all of which still express the basal keratinocyte marker keratin 15. In contrast, epidermal melanocytes remain quiescent under these circumstances. Moreover, the Wnt inhibitors Dickkopf 3 and Wif1 are downregulated upon UVB irradiation in basal keratinocytes, whereas melanocytes continue to express Wnt inhibitors. These findings suggest that there is (a) a suprabasal population, proliferating in the homeostatic state, hence maintaining the integrity of the epidermis, and (b) a basal, usually quiescent keratinocyte population that is induced to massively proliferate upon irradiation. Importantly, the finding that MelSkin responds in a physiological fashion to UVB is of paramount importance in light of the planned clinical application.


Asunto(s)
Queratinocitos/citología , Queratinocitos/efectos de la radiación , Piel/citología , Rayos Ultravioleta , Proteínas Wnt/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Animales , Proliferación Celular/efectos de la radiación , Quimiocinas , Niño , Preescolar , Células Epidérmicas/efectos de la radiación , Humanos , Lactante , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Queratinas/metabolismo , Ratas , Proteínas Represoras/metabolismo , Piel Artificial , Proteínas Wnt/metabolismo , Cicatrización de Heridas/efectos de la radiación
19.
Pediatr Surg Int ; 34(2): 129-135, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29124400

RESUMEN

AIMS AND OBJECTIVES: Vascularized bio-engineered human dermo-epidermal skin substitutes (vascDESS) hold promise for treating burn patients, including those with severe full-thickness wounds. We have previously shown that vascDESS promote wound healing by enhanced influx of macrophages and granulocytes. Immediately following transplantation, macrophages infiltrate the graft and differentiate into a pro-inflammatory (M1) or a pro-healing M2 phenotype. The aim of this study was to characterize the activation state of macrophages infiltrating skin transplants at distinct time points following transplantation. METHODS: Keratinocytes and the stromal vascular fraction (SVF) were derived from human skin or adipose tissue, respectively. Human SVF containing both endothelial and mesenchymal/stromal cells was used to generate vascularized dermal component in vitro, which was subsequently covered with human keratinocytes. Finally, vascDESS were transplanted on the back of immuno-incompetent rats, excised, and analyzed after 1 and 3 weeks using immunohistological techniques. RESULTS: A panel of markers of macrophage M1 (nitric oxide synthase: iNOS) and M2 (CD206) subclass was used. All skin grafts were infiltrated by both M1 and M2 rat macrophages between 1-3 weeks post-transplantation. CD68 (PG-M1) was used as a pan-macrophage marker. The number of CD68+CD206+ M2-polarized macrophages was higher in 3-week transplants as compared to early-stage transplants (1 week). In contrast, the number of CD68+iNOS+ M1 cells was markedly decreased in later stages in vivo. CONCLUSIONS: Macrophages exhibit a heterogeneous and temporally regulated polarization during skin wound healing. Our results suggest that the phenotype of macrophages changes during healing from a more pro-inflammatory (M1) profile in early stages after injury, to a less inflammatory, pro-healing (M2) phenotype in later phases in vivo.


Asunto(s)
Tejido Adiposo/citología , Dermis/citología , Células Epidérmicas , Queratinocitos/citología , Macrófagos/citología , Trasplante de Piel/métodos , Ingeniería de Tejidos/métodos , Adolescente , Animales , Biomarcadores/metabolismo , Células Cultivadas , Niño , Preescolar , Dermis/metabolismo , Epidermis/metabolismo , Humanos , Lactante , Queratinocitos/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Células Madre Mesenquimatosas/citología , Modelos Animales , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Ratas , Receptores de Superficie Celular/metabolismo , Piel Artificial , Cicatrización de Heridas
20.
Pediatr Surg Int ; 34(2): 237-244, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29039047

RESUMEN

AIMS AND OBJECTIVES: The use of autologous tissue-engineered skin substitutes is a promising approach to cover large skin defects in patients. Preclinical investigation is pivotal to test and improve the quality of these bio-engineered substitutes. In the skin, the epidermis, formed mainly by keratinocytes, provides the first physical barrier protecting from the environment. Proper keratinocyte differentiation and, thus, formation of a stratified epidermis is essential for this function. Keratins, the main structural support of keratinocytes, play a vital role regarding differentiation of keratinocytes. Here, we examined the expression pattern of a recently described keratinocyte differentiation marker, namely Keratin 24, in our skin substitutes. MATERIALS AND METHODS: Human epidermal keratinocytes, melanocytes, dermal fibroblasts, palmar fibroblasts or sweat gland cells were used to prepare skin substitutes. Fibroblast-containing collagen hydrogels were prepared, and keratinocytes or sweat gland cells and melanocytes were seeded onto the hydrogels. The generated tissue-engineered dermo-epidermal skin analogs were transplanted onto full-thickness skin wounds created on the back of immuno-incompetent rats. The skin substitutes were excised at different time points and histologically examined with regard to Keratin 24 expression. RESULTS: We observed the expression of Keratin 24 in keratinocytes of the upper stratum spinosum of the epidermis. In particular, we observed an intensified expression of Keratin 24 13 weeks after transplantation compared to 4 weeks after transplantation. Importantly, we noticed a markedly higher presence of Keratin 24 in more spinous layers if we used palmar fibroblasts or sweat gland cells in our skin substitutes compared non-palmar fibroblasts or epidermal keratinocytes. CONCLUSION: Our observations prove that the keratinocyte differentiation marker Keratin 24 is expressed in our dermo-epidermal skin substitutes in a normal pattern. This highlights that our bio-engineered skin analogs mature and reach homeostasis in an in vivo assay. These findings harbor favorable implications regarding future clinical application.


Asunto(s)
Dermis/trasplante , Epidermis/trasplante , Regulación de la Expresión Génica , Queratinas Tipo I/genética , Trasplante de Piel/métodos , Ingeniería de Tejidos/métodos , Heridas y Lesiones/genética , Adolescente , Animales , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Dermis/citología , Células Epidérmicas , Femenino , Humanos , Lactante , Queratinas Tipo I/biosíntesis , Masculino , ARN/genética , Ratas , Piel/lesiones , Piel Artificial , Heridas y Lesiones/patología , Heridas y Lesiones/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...