Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Exp Dermatol ; 33(1): e14998, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38284186

RESUMEN

In photoaged human skin, type I collagen fragmentation impairs dermal extracellular matrix (ECM) integrity, resulting in collapsed/contracted fibroblasts with reduced type I procollagen synthesis. Injections of cross-linked hyaluronic acid (CL-HA) reverse these deleterious changes. To investigate the time course and effects of biochemical changes induced by injected CL-HA, particularly whether fibroblast activation leads to accumulation/deposition of dermal collagen, we injected CL-HA into photoaged skin of human participants over 60 years-old and performed biochemical/microscopic analyses of skin samples. Beginning 1 week post-injection and lasting 6-9 months, fibroblasts exhibited activation, including increased immunostaining and gene expression of markers of type I collagen synthesis, such as heat shock protein 47 and components of the transforming growth factor-ß pathway. At 1 week post-injection, multiphoton microscopy revealed elongation/stretching of fibroblasts, indicating enhanced dermal mechanical support. At 4 weeks, second-harmonic generation microscopy revealed thick collagen bundles densely packed around pools of injected CL-HA. At 12 months, accumulation of thick collagen bundles was observed and injected CL-HA remained present in substantial amounts. Thus, by occupying space in the dermal ECM, injected CL-HA rapidly and durably enhances mechanical support, stimulating fibroblast elongation and activation, which results in thick, densely packed type I collagen bundles accumulating as early as 4 weeks post-injection and continuing for at least a year. These observations indicate that early and prolonged clinical improvement following CL-HA injection results from space-filling and collagen deposition. As type I collagen has an estimated half-life of 15 years, our data provide the foundations for optimizing the timing/frequency of repeat CL-HA injections.


Asunto(s)
Colágeno Tipo I , Ácido Hialurónico , Humanos , Persona de Mediana Edad , Colágeno Tipo I/metabolismo , Ácido Hialurónico/metabolismo , Colágeno/metabolismo , Piel/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo
2.
PLoS One ; 18(12): e0292791, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38064445

RESUMEN

Collagen is the major structural protein in the skin. Fragmentation and disorganization of the collagen fibrils are the hallmarks of the aged human skin dermis. These age-related alterations of collagen fibrils impair skin structural integrity and make the tissue microenvironment more prone to skin disorders. As the biological function of collagen lies predominantly in its physical properties, we applied atomic force microscopy (AFM) and nanoindentation to evaluate the physical properties (surface roughness, stiffness, and hardness) of dermal collagen in young (25±5 years, N = 6) and aged (75±6 years, N = 6) healthy sun-protected hip skin. We observed that in the aged dermis, the surface of collagen fibrils was rougher, and fiber bundles were stiffer and harder, compared to young dermal collagen. Mechanistically, the age-related elevation of matrix metalloproteinase-1 (MMP-1) and advanced glycation end products (AGEs) are responsible for rougher and stiffer/harder dermal collagen, respectively. Analyzing the physical properties of dermal collagen as a function of age revealed that alterations of the physical properties of collagen fibrils changed with age (22-89 years, N = 18). We also observed that the reticular dermis is rougher and mechanically stiffer and harder compared to the papillary dermis in human skin. These data extend the current understanding of collagen beyond biological entities to include biophysical properties.


Asunto(s)
Colágeno , Piel , Humanos , Colágeno/metabolismo , Piel/metabolismo , Dermis/metabolismo , Matriz Extracelular/metabolismo , Epidermis/metabolismo
3.
J Cell Commun Signal ; 17(3): 523-529, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37067763

RESUMEN

This article summarizes important molecular mechanisms that drive aging in human skin from the perspective of dermal fibroblasts. The dermis comprises the bulk of the skin and is largely composed of a collagen-rich extracellular matrix (ECM). The dermal ECM provides mechanical strength, resiliency, and an environment that supports the functions of ibroblasts and other types of dermal cells. Fibroblasts produce the dermal ECM and maintain its homeostasis. Fibroblasts attach to the ECM and this attachment controls their morphology and function. During aging, the ECM undergoes gradual degradation that is nitiated by matrix metalloproteinases (MMPs). This degradation alters mechanical forces within the dermal ECM and disrupts he interactions between fibroblasts and the ECM thereby generating an aged fibroblast phenotype. This aged fibroblast phenotype is characterized by collapsed morphology, altered mechanosignaling, induction of CCN1, and activation of transcription factor AP-1, with consequent upregulation of target genes including MMPs and pro-inflammatory mediators. The TGF-beta pathway coordinately regulates ECM production and turnover. Altered mechanical forces, due to ECM fragmentation, down-regulate the type II TGF-beta receptor, thereby reducing ECM production and further increasing ECM breakdown. Thus, dermal aging involves a feed-forward process that reinforces the aged dermal fibroblast phenotype and promotes age-related dermal ECM deterioration. As discussed in the article, the expression of the aged dermal fibroblast phenotype involves both adaptive and cell-autonomous mechanisms.

4.
J Invest Dermatol ; 143(9): 1700-1707.e1, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36914001

RESUMEN

Fragmentation, disorganization, and depletion of the collagen-rich dermal extracellular matrix are hallmarks of aged human skin. These deleterious alterations are thought to critically mediate many of the prominent clinical attributes of aged skin, including thinning, fragility, impaired wound healing, and a propensity for carcinoma. Matrix metalloproteinase-1 (MMP1) initiates the cleavage of collagen fibrils and is significantly increased in dermal fibroblasts in aged human skin. To investigate the role of elevated MMP1 in skin aging, we generated a conditional bitransgenic mouse (type I collagen alpha chain 2; human MMP1 [Col1a2;hMMP1]) that expresses full-length, catalytically active hMMP1 in dermal fibroblasts. hMMP1 expression is activated by a tamoxifen-inducible Cre recombinase that is driven by the Col1a2 promoter and upstream enhancer. Tamoxifen induced hMMP1 expression and activity throughout the dermis Col1a2:hMMP1 mice. At 6 months of age, Col1a2;hMMP1 mice displayed loss and fragmentation of dermal collagen fibrils, which was accompanied by many of the features of aged human skin, such as contracted fibroblast morphology, reduced collagen production, increased expression of multiple endogenous MMPs, and proinflammatory mediators. Interestingly, Col1a2;hMMP1 mice displayed substantially increased susceptibility to skin papilloma development. These data demonstrate that fibroblast expression of hMMP1 is a critical mediator of dermal aging and creates a dermal microenvironment that promotes keratinocyte tumor development.


Asunto(s)
Papiloma , Envejecimiento de la Piel , Humanos , Animales , Ratones , Anciano , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno/metabolismo , Piel/metabolismo , Envejecimiento de la Piel/genética , Fibroblastos/metabolismo , Células Cultivadas , Microambiente Tumoral
5.
J Cell Commun Signal ; 17(1): 7-11, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36790605

RESUMEN

In celebration of the twentieth anniversary of the inception of the CCN society, and of the first post-Covid-19 live meeting, the executive board of the ICCNS had chosen Nice as the venue for the 11th International workshop on the CCN family of genes. On this occasion participation in the meeting was extended to colleagues from other cell signaling fields who were invited to present both an overview of their work and the future directions of their laboratory. Also, for the first time, the members of the JCCS Editorial Board were invited to participate in a JCCS special session during which all aspects of the journal « life ¼ were addressed and opened to free critical discussion. The scientific presentations and the discussions that followed showed once more that an expansion of the session topics was beneficial to the quality of the meeting and confirmed that the ARBIOCOM project discussed last April in Nice was now on track to be launched in 2023. The participants unanimously welcomed Professor Attramadal's proposition to organize the 2024, 12th International CCN workshop in Oslo, Norway.

6.
JCI Insight ; 7(21)2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36136452

RESUMEN

Activation of TLR4 by its cognate damage-associated molecular patterns (DAMPs) elicits potent profibrotic effects and myofibroblast activation in systemic sclerosis (SSc), while genetic targeting of TLR4 or its DAMPs in mice accelerates fibrosis resolution. To prevent aberrant DAMP/TLR4 activity, a variety of negative regulators evolved to dampen the magnitude and duration of the signaling. These include radioprotective 105 kDa (RP105), a transmembrane TLR4 homolog that competitively inhibits DAMP recognition of TLR4, blocking TLR4 signaling in immune cells. The role of RP105 in TLR4-dependent fibrotic responses in SSc is unknown. Using unbiased transcriptome analysis of skin biopsies, we found that levels of both TLR4 and its adaptor protein MD2 were elevated in SSc skin and significantly correlated with each other. Expression of RP105 was negatively associated with myofibroblast differentiation in SSc. Importantly, RP105-TLR4 association was reduced, whereas TLR4-TLR4 showed strong association in fibroblasts from patients with SSc, as evidenced by PLA assays. Moreover, RP105 adaptor MD1 expression was significantly reduced in SSc skin biopsies and explanted SSc skin fibroblasts. Exogenous RP105-MD1 abrogated, while loss of RP105 exaggerated, fibrotic cellular responses. Importantly, ablation of RP105 in mice was associated with augmented TLR4 signaling and aggravated skin fibrosis in complementary disease models. Thus, we believe RP105-MD1 to be a novel cell-intrinsic negative regulator of TLR4-MD2-driven sustained fibroblast activation, representing a critical regulatory network governing the fibrotic process. Impaired RP105 function in SSc might contribute to persistence of progression of the disease.


Asunto(s)
Esclerodermia Sistémica , Receptor Toll-Like 4 , Ratones , Animales , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Fibrosis , Fibroblastos/metabolismo , Transducción de Señal , Alarminas/metabolismo
7.
J Cell Commun Signal ; 16(3): 421-432, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35060094

RESUMEN

Collagen 17A1 (COL17A1) is a transmembrane structural component of the hemidesmosome that mediate adhesion of keratinocytes to the underlying membrane. Recent work in mouse showed that COL17A1 deficiency leads to premature skin aging. Although the role COL17A1 in skin aging is becoming recognized in mouse models, its connection to human skin natural aging/photoaging/ultraviolet (UV)-irradiated human skin has received little attention. To determine COL17A1 expression in naturally aged and photoaged as well as acutely UV irradiated human skin, skin samples were obtained from: (1) young (N = 10, 26.7±1.3 years) and aged (N = 10, 84.0 ± 1.7 years) sun-protected buttock skin; (2) photoaged extensor forearm and subject matched sun-protected underarm skin (N = 6, 56.0 ± 3.4 years); (3) solar-simulated UV-irradiated buttock skin (N = 6, 51.2 ± 3.6 years). COL17A1 levels were determined by immunohistology and RT-PCR, and the potential role of COL17A1 in epidermal aging was investigated by immunostaining of the marker for interfollicular epidermal stem cells and keratinocytes proliferation. We found that COL17A1 is specifically expressed in interfollicular epidermal stem cell niches, and that significantly reduced in naturally aged, photoaged, and acute UV-irradiated human skin in vivo. COL17A1 is identified as keratinocyte-specific collagen, and UV irradiation significantly downregulates COL17A1 expression in keratinocytes. Reduced expression of COL17A1 is positively correlated with impaired regeneration of keratinocytes and reduced dermal-epidermal junction as well as thin epidermis in aged human skin (epidermal aging). We also confirmed that keratinocyte-specific integrin ß4 (ITGB4), which interacts with COL17A1, is reduced in aged human skin. Mechanistically, we found that matrix metalloproteinases (MMPs) are responsible for UV-mediated COL17A1 degradation in both in vitro keratinocytes and in vivo mouse skin. These data suggest the possible links between reduced expression of COL17A1 and epidermal aging in human skin.

8.
J Dermatol Sci ; 102(1): 36-46, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33648801

RESUMEN

BACKGROUND: Aged human skin is primarily attributable to the loss of collagen. Hepatocyte growth factor (HGF) acts as an anti-fibrotic factor by suppression of collagen production. In aged human skin, HGF is elevated in dermal fibroblasts and thus contributes to dermal aging (thin dermis) by suppression of collagen production. OBJECTIVE: We aimed to investigate the underlying mechanisms of age-related elevation of HGF expression. METHODS: Collagen fibrils in the aged skin dermis are fragmented and disorganized, which impairs collagen-fibroblast interaction, resulting in reduced fibroblast spreading and size. To explore the connection between reduced dermal fibroblast size and age-related elevation of HGF expression, we manipulate dermal fibroblast size, and cell-size dependent regulation of HGF was investigated by laser capture microdissection, immunostaining, capillary electrophoresis immunoassay, and quantitative RT-PCR. RESULTS: We found that reduced fibroblast size is responsible for age-related elevation of HGF expression. Further investigation indicated that cell size-dependent upregulation of HGF expression was mediated by impeded YAP/TAZ nuclear translocation and their target gene, CCN2. Conversely, restoration of dermal fibroblast size rapidly reversed cell-size-dependent upregulation of HGF in a YAP/TAZ-dependent manner. Finally, we confirmed that elevated HGF expression is accompanied by the reduced expression of YAP/TAZ and CCN2 in the aged human skin in vivo. CONCLUSION: Age-related elevation of HGF is driven by the reduction of fibroblast size in a YAP/TAZ/CCN2 axis-dependent manner. These data reveal a novel mechanism by which reduction of fibroblast size upregulates HGF expression, which in turn contributes to loss of collagen, a prominent feature of aged human skin.


Asunto(s)
Fibroblastos/patología , Factor de Crecimiento de Hepatocito/genética , Envejecimiento de la Piel/genética , Piel/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano de 80 o más Años , Biopsia , Tamaño de la Célula , Células Cultivadas , Colágeno/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cultivo Primario de Células , Transducción de Señal/genética , Piel/citología , Piel/metabolismo , Factores de Transcripción/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Regulación hacia Arriba , Proteínas Señalizadoras YAP , Adulto Joven
9.
J Invest Dermatol ; 141(4S): 1007-1016, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32800875

RESUMEN

The aging process deleteriously alters the structure and function of dermal collagen. These alterations result in thinning, fragility, wrinkles, laxity, impaired wound healing, and a microenvironment conducive to cancer. However, the key factors responsible for these changes have not been fully elucidated, and relevant models for the study of skin aging progression are lacking. CCN1, a secreted extracellular matrix‒associated matricellular protein, is elevated in dermal fibroblasts in aged human skin. Toward constructing a mouse model to study the key factors involved in skin-aging progression, we demonstrate that transgenic mice, with selective expression of CCN1 in dermal fibroblasts (COL1A2-CCN1), display accelerated skin dermal aging. The aged phenotype in COL1A2-CCN1 mice resembles aged human dermis: the skin is wrinkled and the dermis is thin and composed of loose, disorganized, and fragmented collagen fibrils. These dermal alterations reflect reduced production of collagen due to impaired TGFß signaling and increased expression of matrix metalloproteinases driving the induction of c-Jun/activator protein-1. Importantly, similar mechanisms drive human dermal aging. Taken together, the data demonstrate that elevated expression of CCN1 by dermal fibroblasts functions as a key mediator of dermal aging. The COL1A2-CCN1 mouse model provides a novel tool for understanding and studying the mechanisms of skin aging and age-related skin disorders.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Dermis/patología , Fibroblastos/patología , Envejecimiento de la Piel , Animales , Células Cultivadas , Colágeno/metabolismo , Colágeno Tipo I/genética , Proteína 61 Rica en Cisteína/genética , Dermis/citología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Modelos Animales , Cultivo Primario de Células , Regiones Promotoras Genéticas/genética , Regulación hacia Arriba
10.
Plast Reconstr Surg ; 147(1S-2): 43S-49S, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33347074

RESUMEN

BACKGROUND: Dermal injection of chemically cross-linked hyaluronic acid (CL-HA) is a common procedure to smooth wrinkles and add fullness to the face. Due to its physical properties, CL-HA both fills space and exerts mechanical forces within the dermis. Dermal fibroblasts produce the collagen-rich extracellular matrix (ECM), which comprises the bulk of skin. Attachment to the ECM allows fibroblasts to achieve a stretched, morphology, which confers a functional phenotype that maintains collagen production. In aged/photoaged skin, collagen fibril fragmentation impairs fibroblast attachment, resulting in a collapsed morphology and reduced collagen production. This article describes investigations of the impact of CL-HA injection on fibroblast morphology and function in the aged/photoaged human skin. METHODS: Fifty-three subjects, age 70 years or older, received a single injection of saline (vehicle control) and CL-HA (0.5 ml each) in separate adjacent skin sites on photodamaged forearm or sun-protected buttock skin. Full-thickness punch biopsies were obtained from injected skin sites at various times and analyzed for molecular and cellular changes. RESULTS: Injected CL-HA forms discreet pockets that localize to areas of the dermis that contain fragmented, loosely organized collagen fibrils. These CL-HA pockets fill space and apply mechanical forces on adjacent ECM that induce stretching of fibroblasts. This stretching is associated with increased collagen gene expression and deposition of mature collagen fibril bundles, which resemble those observed in young skin. CONCLUSIONS: CL-HA injected into aged/photoaged human dermis acts by both filling space and inducing production of collagen by dermal fibroblasts. Deposition of mature collagen, which remains in the skin for decades, likely confers long-term benefits. Reduced collagen production in aged/photoaged skin is an adaptive response of fibroblasts to ECM fragmentation, rather than inherent cellular aging mechanisms.


Asunto(s)
Rellenos Dérmicos/administración & dosificación , Dermis/efectos de los fármacos , Ácido Hialurónico/administración & dosificación , Rejuvenecimiento , Envejecimiento de la Piel/efectos de los fármacos , Anciano , Colágeno/metabolismo , Reactivos de Enlaces Cruzados/química , Rellenos Dérmicos/química , Dermis/citología , Dermis/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ácido Hialurónico/química , Inyecciones Intradérmicas , Masculino , Resultado del Tratamiento
11.
J Invest Dermatol ; 140(12): 2515-2524.e3, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32407715

RESUMEN

Fibroblasts produce collagens and other proteins that form the bulk of the extracellular matrix (ECM) in connective tissues. Emerging data point to functional heterogeneity of fibroblasts. However, the lack of subtype-specific markers hinders our understanding of the different roles of fibroblasts in ECM biology, wound healing, diseases, and aging. We have investigated the utility of the cell surface protein CD26 to identify functionally distinct fibroblast subpopulations in human skin. Using flow cytometry and immunohistology, we found that CD26, in combination with the cell surface glycoprotein CD90, identifies a distinct subpopulation of cells, which express relatively high levels of COL1A1, a hallmark of fibroblasts. Importantly, the population of CD26+ fibroblasts is selectively increased after wounding of human skin. These cells account for the majority of COL1A1 expression during the ECM remodeling phase of healing. The proportion of CD26+ fibroblasts in the skin of young and aged individuals is similar, indicating that the loss of collagen production during aging does not involve selective reduction of CD26+ fibroblasts. In culture, the majority of freshly isolated CD26- fibroblasts gain expression of CD26+. Taken together, these data provide a foundation for targeting CD26+ fibroblasts to modulate wound healing in human skin.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Fibroblastos/metabolismo , Piel/metabolismo , Cicatrización de Heridas/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Separación Celular , Células Cultivadas , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Citometría de Flujo , Humanos , Persona de Mediana Edad , Cultivo Primario de Células , Piel/citología , Envejecimiento de la Piel/fisiología , Antígenos Thy-1/metabolismo , Adulto Joven
12.
Matrix Biol Plus ; 8: 100041, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33543036

RESUMEN

Human skin is composed of the cell-rich epidermis, the extracellular matrix (ECM) rich dermis, and the hypodermis. Within the dermis, a dense network of ECM proteins provides structural support to the skin and regulates a wide variety of signaling pathways which govern cell proliferation and other critical processes. Both intrinsic aging, which occurs steadily over time, and extrinsic aging (photoaging), which occurs as a result of external insults such as solar radiation, cause alterations to the dermal ECM. In this study, we utilized both quantitative and global proteomics, alongside single harmonic generation (SHG) and two-photon autofluorescence (TPAF) imaging, to assess changes in dermal composition during intrinsic and extrinsic aging. We find that both intrinsic and extrinsic aging result in significant decreases in ECM-supporting proteoglycans and structural ECM integrity, evidenced by decreasing collagen abundance and increasing fibril fragmentation. Intrinsic aging also produces changes distinct from those produced by photoaging, including reductions in elastic fiber and crosslinking enzyme abundance. In contrast, photoaging is primarily defined by increases in elastic fiber-associated protein and pro-inflammatory proteases. Changes associated with photoaging are evident even in young (mid 20s) sun-exposed forearm skin, indicating that proteomic evidence of photoaging is present decades prior to clinical signs of photoaging. GO term enrichment revealed that both intrinsic aging and photoaging share common features of chronic inflammation. The proteomic data has been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD015982.

13.
J Am Acad Dermatol ; 81(2): 480-488, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30954583

RESUMEN

BACKGROUND: Exposure to the sun causes premature skin aging, known as photoaging. Clinical features of photoaging vary widely among individuals. In one form, skin appears thin with telangiectasia, and in another form, skin appears thickened with coarse wrinkles. Etiologic, clinical, and therapeutic distinctions among different forms of photoaging remain largely unknown. OBJECTIVE: To characterize the clinical, histologic, and molecular features of hypertrophic and atrophic photoaging. METHODS: In total, 53 individuals were clinically classified as having primarily atrophic or hypertrophic photoaging or neither (controls). Participants' demographic and sun exposure-related lifestyle data were captured by questionnaire. Fifteen clinical features of participants were qualitatively or quantitively scored. Facial biopsies were analyzed for gene expression and histologic characteristics. RESULTS: Actinic and seborrheic keratosis, telangiectasia, and prior incidence of skin cancers were statistically significantly greater and photoaging scale severity, coarse wrinkles, thickness, and sallowness were significantly reduced in atrophic versus hypertrophic groups. Histology also revealed significantly less elastotic material in atrophic photoaging. Gene expression of matrix metalloproteinases and collagens did not differ between the 2 forms of photoaging. LIMITATIONS: The study was not designed to identify other possible subtypes of photoaging. CONCLUSION: Systematic, categorical, and quantitative clinical and histologic assessments distinguish atrophic and hypertrophic photoaging.


Asunto(s)
Carcinoma Basocelular/epidemiología , Carcinoma de Células Escamosas/epidemiología , Envejecimiento de la Piel/genética , Envejecimiento de la Piel/patología , Neoplasias Cutáneas/epidemiología , Piel/metabolismo , Piel/patología , Anciano , Anciano de 80 o más Años , Atrofia/genética , Atrofia/patología , Biopsia , Colágeno/genética , Cara , Femenino , Expresión Génica , Humanos , Hipertrofia/genética , Hipertrofia/patología , Incidencia , Queratosis Actínica/epidemiología , Queratosis Seborreica/epidemiología , Estilo de Vida , Masculino , Metaloproteinasas de la Matriz/genética , Persona de Mediana Edad , Fenotipo , Piel/efectos de la radiación , Envejecimiento de la Piel/efectos de la radiación , Encuestas y Cuestionarios , Telangiectasia/epidemiología , Telangiectasia/patología , Rayos Ultravioleta/efectos adversos
14.
Exp Dermatol ; 28(8): 914-921, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-29957839

RESUMEN

Fragmentation of collagen fibrils and aberrant elastic material (solar elastosis) in the dermal extracellular matrix (ECM) is among the most prominent features of photodamaged human skin. These alterations impair the structural integrity and create a dermal microenvironment prone to skin disorders. The objective of this study was to determine the physical properties (surface roughness, stiffness and hardness) of the dermal ECM in photodamaged and subject-matched sun-protected human skin. Skin samples were sectioned and analysed by histology, atomic force microscopy and nanoindentation. Dermal ECM collagen fibrils were more disorganized (ie, rougher surface), and the dermal ECM was stiffer and harder, in photodamaged forearm, compared to sun-protected underarm skin. Cleavage of collagen fibrils in sun-protected underarm dermis by recombinant human matrix metalloproteinase-1 resulted in rougher collagen fibril surface and reduced dermal stiffness and hardness. Degradation of elastotic material in photodamaged skin by treatment with purified neutrophil elastase reduced stiffness and hardness, without altering collagen fibril surface roughness. Additionally, expression of two members of the lysyl oxidase gene family, which insert cross-links that stiffen and harden collagen fibrils, was elevated in photodamaged forearm dermis. These data elucidate the contributions of fragmented collagen fibrils, solar elastosis and elevated collagen cross-linking to the physical properties of the dermal ECM in photodamaged human skin. This new knowledge extends current understanding of the impact of photodamage on the dermal ECM microenvironment.


Asunto(s)
Colágeno , Dermis/patología , Envejecimiento de la Piel/patología , Estudios de Casos y Controles , Dermis/enzimología , Matriz Extracelular/patología , Dureza , Humanos , Persona de Mediana Edad , Proteína-Lisina 6-Oxidasa/metabolismo , Luz Solar/efectos adversos
15.
Exp Dermatol ; 28(1): 11-18, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30329180

RESUMEN

Systemic sclerosis (SSc) is a complex autoimmune connective tissue disease combining inflammatory, vasculopathic and fibrotic manifestations. Skin features, which give their name to the disease and are considered as diagnostic as well as prognostic markers, have not been thoroughly investigated in terms of therapeutic targets. CCN proteins (CYR61/CCN1, CTGF/CCN2, NOV/CCN3 and WISP1-2-3 as CCN4-5-6) are a family of secreted matricellular proteins implicated in major cellular processes such as cell growth, migration, differentiation. They have already been implicated in key pathophysiological processes of SSc, namely fibrosis, vasculopathy and inflammation. In this review, we discuss the possible implication of CCN proteins in SSc pathogenesis, with a special focus on skin features, and identify the potential actionable CCN targets.


Asunto(s)
Proteínas CCN de Señalización Intercelular/metabolismo , Neovascularización Patológica/metabolismo , Esclerodermia Sistémica/metabolismo , Piel/patología , Envejecimiento , Animales , Autoinmunidad , Proteínas CCN de Señalización Intercelular/genética , Diferenciación Celular , Dermatitis/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Humanos , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/fisiopatología , Piel/irrigación sanguínea , Piel/metabolismo , Pigmentación de la Piel
16.
J Cell Mol Med ; 22(9): 4085-4096, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29888864

RESUMEN

The dermal compartment of skin is primarily composed of collagen-rich extracellular matrix (ECM), which is produced by dermal fibroblasts. In Young skin, fibroblasts attach to the ECM through integrins. During ageing, fragmentation of the dermal ECM limits fibroblast attachment. This reduced attachment is associated with decreased collagen production, a major cause of skin thinning and fragility, in the elderly. Fibroblast attachment promotes assembly of the cellular actin cytoskeleton, which generates mechanical forces needed for structural support. The mechanism(s) linking reduced assembly of the actin cytoskeleton to decreased collagen production remains unclear. Here, we report that disassembly of the actin cytoskeleton results in impairment of TGF-ß pathway, which controls collagen production, in dermal fibroblasts. Cytoskeleton disassembly rapidly down-regulates TGF-ß type II receptor (TßRII) levels. This down-regulation leads to reduced activation of downstream effectors Smad2/Smad3 and CCN2, resulting in decreased collagen production. These responses are fully reversible; restoration of actin cytoskeleton assembly up-regulates TßRII, Smad2/Smad3, CCN2 and collagen expression. Finally, actin cytoskeleton-dependent reduction of TßRII is mediated by induction of microRNA 21, a potent inhibitor of TßRII protein expression. Our findings reveal a novel mechanism that links actin cytoskeleton assembly and collagen expression in dermal fibroblasts. This mechanism likely contributes to loss of TßRII and collagen production, which are observed in aged human skin.


Asunto(s)
Citoesqueleto de Actina/genética , Fibroblastos/metabolismo , Procolágeno/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Adulto , Senescencia Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Cultivo Primario de Células , Procolágeno/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Piel/citología , Piel/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
18.
Cell Commun Signal ; 16(1): 18, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29695252

RESUMEN

BACKGROUND: Transcription factors YAP and TAZ function as the primary mediators of the Hippo pathway. Yet, crosstalk of YAP and TAZ with other signaling pathways remains relatively unexplored. We have explored the impact of YAP and TAZ levels on the TGF-ß/Smad signaling pathway in human skin dermal fibroblasts. METHODS: YAP and TAZ levels in dermal fibroblasts were reduced in dermal fibroblasts by siRNA-mediated knockdown. The effects of YAP and TAZ reduction on TGF-ß/Smad signaling were examined by quantitative real-time PCR, Western analysis, and immunostaining. Luciferase reporter assays and electrophoretic mobility shift assays were conducted to investigate the transcription factor DNA-binding and transcriptional activities. RESULTS: Knockdown of both YAP and TAZ (YAP/TAZ), but not either separately, impaired TGF-ß1-induced Smad3 phosphorylation and Smad3 transcriptional activity, thereby inhibiting the expression of TGF-ß target genes. This reduction by reduced levels of YAP/TAZ results from induction of inhibitory Smad7, which inhibits Smad3 phosphorylation and activity by TGF-ß1. Conversely, prevention of Smad7 induction restores Smad3 phosphorylation and Smad3 transcriptional activity in fibroblasts that have reduced YAP/TAZ. In agreement with these findings, inhibition of YAP/TAZ transcriptional activity, similar to the reduction of YAP/TAZ levels, also significantly induced Smad7 and impaired TGF-ß/Smad signaling. Further investigations revealed that reduced levels of YAP/TAZ led to induction of activator protein-1 (AP-1) activity, Activated AP-1 bound to DNA sequences in the Smad7 gene promoter, and deletion of these AP-1 binding sequences substantially reduced Smad7 promoter reporter activity. CONCLUSION: YAP/TAZ functions in concert with transcription factor AP-1 and Smad7 to regulate TGF-ß signaling, in human dermal fibroblasts. Reduction of YAP/TAZ levels leads to activation of AP-1 activity, which induces Smad7. Smad7 suppresses the TGF-ß pathway.


Asunto(s)
Proteínas Nucleares/metabolismo , Transducción de Señal , Proteína smad7/metabolismo , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Células Cultivadas , Dermis/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína smad7/antagonistas & inhibidores , Proteína smad7/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
19.
J Cell Commun Signal ; 12(1): 35-43, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29455303

RESUMEN

The dermal extracellular matrix (ECM) comprises the bulk of skin and confers strength and resiliency. In young skin, fibroblasts produce and adhere to the dermal ECM, which is composed primarily of type I collagen fibrils. Adherence allows fibroblasts to spread and exert mechanical force on the surrounding ECM. In this state, fibroblasts display a "youthful" phenotype characterized by maintenance of the composition and structural organization of the dermal ECM. During aging, fibroblast-ECM interactions become disrupted due to fragmentation of collagen fibrils. This disruption causes loss of fibroblast spreading and mechanical force, which inextricably lead to an "aged" phenotype; fibroblasts synthesize less ECM proteins and more matrix-degrading metalloproteinases. This imbalance of ECM homeostasis further drives collagen fibril fragmentation in a self-perpetuating cycle. This article summarizes age-related changes in the dermal ECM and the mechanisms by which these changes alter the interplay between fibroblasts and their extracellular matrix microenvironment that drive the aging process in human skin.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...