Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioeng Transl Med ; 7(1): e10247, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35111948

RESUMEN

Atopic dermatitis (AD), driven by interleukins (IL-4/IL-13), is a chronic inflammatory skin disease characterized by intensive pruritus. However, it is unclear how immune signaling and sensory response pathways cross talk with each other. We differentiated itch sensory neuron-like cells (ISNLCs) from iPSC lines. These ISNLCs displayed neural markers and action potentials and responded specifically to itch-specific stimuli. These ISNLCs expressed receptors specific for IL-4/IL-13 and were activated directly by the two cytokines. We successfully innervated these ISNLCs into full thickness human skin constructs. These innervated skin grafts can be used in clinical applications such as wound healing. Moreover, the availability of such innervated skin models will be valuable to develop drugs to treat skin diseases such as AD.

2.
Sci Rep ; 10(1): 4123, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32139717

RESUMEN

Drug screening studies for inflammatory skin diseases are currently performed using model systems that only partially recapitulate human diseased skin. Here, we developed a new strategy to incorporate T cells into human 3D skin constructs (HSCs), which enabled us to closely monitor and quantitate T cell responses. We found that the epidermis promotes the activation and infiltration of T cells into the skin, and provides a directional cue for their selective migration towards the epidermis. We established a psoriatic HSC (pHSC) by incorporating polarized Th1/Th17 cells or CCR6+CLA+ T cells derived from psoriasis patients into the constructs. These pHSCs showed a psoriatic epidermal phenotype and characteristic cytokine profiles, and responded to various classes of psoriasis drugs, highlighting the potential utility of our model as a drug screening platform. Taken together, we developed an advanced immunocompetent 3D skin model to investigate epidermal-T cell interactions and to understand the pathophysiology of inflammatory skin diseases in a human-relevant and patient-specific context.


Asunto(s)
Psoriasis/inmunología , Piel/citología , Piel/metabolismo , Células Cultivadas , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/citología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Queratinocitos/citología , Queratinocitos/inmunología , Queratinocitos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CCR6/genética , Receptores CCR6/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/inmunología , Células TH1/metabolismo , Células Th17/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(52): 26846-26852, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31818947

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is a severe inherited skin disorder caused by mutations in the COL7A1 gene encoding type VII collagen (C7). The spectrum of severity depends on the type of mutation in the COL7A1 gene. C7 is the major constituent of anchoring fibrils (AFs) at the basement membrane zone (BMZ). Patients with RDEB lack functional C7 and have severely impaired dermal-epidermal stability, resulting in extensive blistering and open wounds on the skin that greatly affect the patient's quality of life. There are currently no therapies approved for the treatment of RDEB. Here, we demonstrated the correction of mutations in exon 19 (c.2470insG) and exon 32 (c.3948insT) in the COL7A1 gene through homology-directed repair (HDR). We used the clustered regulatory interspaced short palindromic repeats (CRISPR) Cas9-gRNAs system to modify induced pluripotent stem cells (iPSCs) derived from patients with RDEB in both the heterozygous and homozygous states. Three-dimensional human skin equivalents (HSEs) were generated from gene-corrected iPSCs, differentiated into keratinocytes (KCs) and fibroblasts (FBs), and grafted onto immunodeficient mice, which showed normal expression of C7 at the BMZ as well as restored AFs 2 mo postgrafting. Safety assessment for potential off-target Cas9 cleavage activity did not reveal any unintended nuclease activity. Our findings represent a crucial advance for clinical applications of innovative autologous stem cell-based therapies for RDEB.

4.
Nat Commun ; 9(1): 5301, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30546011

RESUMEN

Human skin constructs (HSCs) have the potential to provide an effective therapy for patients with significant skin injuries and to enable human-relevant drug screening for skin diseases; however, the incorporation of engineered skin appendages, such as hair follicles (HFs), into HSCs remains a major challenge. Here, we demonstrate a biomimetic approach for generation of human HFs within HSCs by recapitulating the physiological 3D organization of cells in the HF microenvironment using 3D-printed molds. Overexpression of Lef-1 in dermal papilla cells (DPC) restores the intact DPC transcriptional signature and significantly enhances the efficiency of HF differentiation in HSCs. Furthermore, vascularization of hair-bearing HSCs prior to engraftment allows for efficient human hair growth in immunodeficient mice. The ability to regenerate an entire HF from cultured human cells will have a transformative impact on the medical management of different types of alopecia, as well as chronic wounds, which represent major unmet medical needs.


Asunto(s)
Alopecia/terapia , Dermis/citología , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/trasplante , Ingeniería de Tejidos/métodos , Alopecia/patología , Animales , Biomimética , Diferenciación Celular , Células Cultivadas , Folículo Piloso/citología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factor de Unión 1 al Potenciador Linfoide/biosíntesis , Masculino , Ratones , Ratones Desnudos , Trasplante Heterólogo
5.
Biomaterials ; 166: 96-108, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29549768

RESUMEN

Wounds in the fetus can heal without scarring. Consequently, biomaterials that attempt to recapitulate the biophysical and biochemical properties of fetal skin have emerged as promising pro-regenerative strategies. The extracellular matrix (ECM) protein fibronectin (Fn) in particular is believed to play a crucial role in directing this regenerative phenotype. Accordingly, Fn has been implicated in numerous wound healing studies, yet remains untested in its fibrillar conformation as found in fetal skin. Here, we show that high extensional (∼1.2 ×105 s-1) and shear (∼3 ×105 s-1) strain rates in rotary jet spinning (RJS) can drive high throughput Fn fibrillogenesis (∼10 mL/min), thus producing nanofiber scaffolds that are used to effectively enhance wound healing. When tested on a full-thickness wound mouse model, Fn nanofiber dressings not only accelerated wound closure, but also significantly improved tissue restoration, recovering dermal and epidermal structures as well as skin appendages and adipose tissue. Together, these results suggest that bioprotein nanofiber fabrication via RJS could set a new paradigm for enhancing wound healing and may thus find use in a variety of regenerative medicine applications.


Asunto(s)
Materiales Biocompatibles , Fibronectinas , Nanofibras , Cicatrización de Heridas , Administración Cutánea , Animales , Materiales Biocompatibles/química , Fibronectinas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Nanofibras/química , Piel/efectos de los fármacos , Piel/patología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos
6.
Exp Biol Med (Maywood) ; 242(17): 1657-1668, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28592171

RESUMEN

Many diseases, as well as side effects of drugs, manifest themselves through skin symptoms. Skin is a complex tissue that hosts various specialized cell types and performs many roles including physical barrier, immune and sensory functions. Therefore, modeling skin in vitro presents technical challenges for tissue engineering. Since the first attempts at engineering human epidermis in 1970s, there has been a growing interest in generating full-thickness skin constructs mimicking physiological functions by incorporating various skin components, such as vasculature and melanocytes for pigmentation. Development of biomimetic in vitro human skin models with these physiological functions provides a new tool for drug discovery, disease modeling, regenerative medicine and basic research for skin biology. This goal, however, has long been delayed by the limited availability of different cell types, the challenges in establishing co-culture conditions, and the ability to recapitulate the 3D anatomy of the skin. Recent breakthroughs in induced pluripotent stem cell (iPSC) technology and microfabrication techniques such as 3D-printing have allowed for building more reliable and complex in vitro skin models for pharmaceutical screening. In this review, we focus on the current developments and prevailing challenges in generating skin constructs with vasculature, skin appendages such as hair follicles, pigmentation, immune response, innervation, and hypodermis. Furthermore, we discuss the promising advances that iPSC technology offers in order to generate in vitro models of genetic skin diseases, such as epidermolysis bullosa and psoriasis. We also discuss how future integration of the next generation human skin constructs onto microfluidic platforms along with other tissues could revolutionize the early stages of drug development by creating reliable evaluation of patient-specific effects of pharmaceutical agents. Impact statement Skin is a complex tissue that hosts various specialized cell types and performs many roles including barrier, immune, and sensory functions. For human-relevant drug testing, there has been a growing interest in building more physiological skin constructs by incorporating different skin components, such as vasculature, appendages, pigment, innervation, and adipose tissue. This paper provides an overview of the strategies to build complex human skin constructs that can faithfully recapitulate human skin and thus can be used in drug development targeting skin diseases. In particular, we discuss recent developments and remaining challenges in incorporating various skin components, availability of iPSC-derived skin cell types and in vitro skin disease models. In addition, we provide insights on the future integration of these complex skin models with other organs on microfluidic platforms as well as potential readout technologies for high-throughput drug screening.


Asunto(s)
Descubrimiento de Drogas/métodos , Procedimientos Analíticos en Microchip/métodos , Microfluídica/métodos , Medicina Regenerativa/métodos , Enfermedades de la Piel/patología , Piel/metabolismo , Ingeniería de Tejidos/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Dispositivos Laboratorio en un Chip
7.
Methods Mol Biol ; 1235: 147-64, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25388393

RESUMEN

The epidermis and associated appendages of the skin represent a multi-lineage tissue that is maintained by perpetual rounds of renewal. During homeostasis, turnover of epidermal lineages is achieved by input from regionalized keratinocytes stem or progenitor populations with little overlap from neighboring niches. Over the last decade, molecular markers selectively expressed by a number of these stem or progenitor pools have been identified, allowing for the isolation and functional assessment of stem cells and genetic lineage tracing analysis within intact skin. These advancements have led to many fundamental observations about epidermal stem cell function such as the identification of their progeny, their role in maintenance of skin homeostasis, or their contribution to wound healing. In this chapter, we provide a methodology to identify and isolate epidermal stem cells and to assess their functional role in their respective niche. Furthermore, recent evidence has shown that the microenvironment also plays a crucial role in stem cell function. Indeed, epidermal cells are under the influence of surrounding fibroblasts, adipocytes, and sensory neurons that provide extrinsic signals and mechanical cues to the niche and contribute to skin morphogenesis and homeostasis. A better understanding of these microenvironmental cues will help engineer in vitro experimental models with more relevance to in vivo skin biology. New approaches to address and study these environmental cues in vitro will also be addressed.


Asunto(s)
Separación Celular/métodos , Células Epidérmicas , Queratinocitos/citología , Células Madre/citología , Animales , Técnicas de Cultivo de Célula/métodos , Ensayo de Unidades Formadoras de Colonias/métodos , Citometría de Flujo/métodos , Ratones , Microdisección/métodos , Técnicas de Cultivo de Tejidos/métodos , Ingeniería de Tejidos/métodos
8.
Cell Rep ; 3(6): 1759-65, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23727240

RESUMEN

In mammalian skin, Merkel cells are mechanoreceptor cells that are required for the perception of gentle touch. Recent evidence indicates that mature Merkel cells descend from the proliferative layer of skin epidermis; however, the stem cell niche for Merkel cell homeostasis has not been reported. Here, we provide genetic evidence for maintenance of mature Merkel cells during homeostasis by Krt17+ stem cells located in epidermal touch domes of hairy skin and in the tips of the rete ridges of glabrous skin. Lineage tracing analysis indicated that the entire pool of mature Merkel cells is turned over every 7-8 weeks in the adult epidermis and that Krt17+ stem cells also maintain squamous differentiation in the touch dome and in glabrous skin. Finally, selective genetic ablation of Krt17+ touch-dome keratinocytes indicates that these cells, and not mature Merkel cells, are primarily responsible for maintaining innervation of the Merkel cell-neurite complex.


Asunto(s)
Células Epidérmicas , Células de Merkel/citología , Nicho de Células Madre/fisiología , Tacto/fisiología , Animales , Humanos , Mecanorreceptores/citología , Ratones , Ratones Transgénicos , Transducción de Señal , Piel/inervación
9.
BMC Dermatol ; 13: 2, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23343495

RESUMEN

BACKGROUND: Sebaceous glands are components of the skin essential for its normal lubrication by the production of sebum. This contributes to skin health and more importantly is crucial for the skin barrier function. A mechanistic understanding of sebaceous gland cells growth and differentiation has lagged behind that for keratinocytes, partly because of a lack of an in vitro model that can be used for experimental manipulation. METHODS: We have developed an in vitro culture model to isolate and grow primary human sebocytes without transformation that display functional characteristics of sebocytes. We used this novel method to probe the effect of Transforming Growth Factor ß (TGFß) signaling on sebocyte differentiation, by examining the expression of genes involved in lipogenesis upon treatment with TGFß1. We also repressed TGFß signaling through knockdown of the TGFß Receptor II to address if the effect of TGFß activation is mediated via canonical Smad signal transduction. RESULTS: We find that activation of the TGFß signaling pathway is necessary and sufficient for maintaining sebocytes in an undifferentiated state. The presence of TGFß ligand triggered decreased expression in genes required for the production of characteristics sebaceous lipids and for sebocyte differentiation such as FADS2 and PPARγ, thereby decreasing lipid accumulation through the TGFß RII-Smad2 dependent pathway. CONCLUSION: TGFß signaling plays an essential role in sebaceous gland regulation by maintaining sebocytes in an undifferentiated state. This data was generated using a novel method for human sebocyte culture, which is likely to prove generally useful in investigations of sebaceous gland growth and differentiation. These findings open a new paradigm in human skin biology with important implications for skin therapies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Lipogénesis/fisiología , Glándulas Sebáceas/citología , Glándulas Sebáceas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Mama/citología , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Cara , Fibronectinas/metabolismo , Humanos , Lactante , Cuero Cabelludo/citología , Transducción de Señal , Tórax/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...